250
Monographs on Pathology of Laboratory Animals Sponsored by the International Life Sciences Institute Editorial Board J. D. Burek, West Point· J. S. Campbell, Ottawa C. C. Capen, Columbus . A. Cardesa, Barcelona RG.Carison, Kalamazoo· D. de Paola, Rio de Janeiro G. Della Porta, Milan . J. L. Emerson, Atlanta F. M. Garner, Rockville . L. Golberg, Research Triangle Park H. C. Grice, N epean . C. C. Harris, Bethesda . R Hess, Basel C. F. Hollander, Rijswijk· G. H. Hottendorf, Syracuse RD. Hunt, Southborough . T. C. Jones, Southborough Y. Konishi, Nara . D. Krewski, Ottawa· R Kroes, Bilthoven H. Luginbuhl, Bern . U. Mohr, Hannover . P. Olsen, Soborg J. A. Popp, Research Triangle Park· J. R Schenken, Omaha R A. Squire, Baltimore· J. Sugar, Budapest S. Takayama, Tokyo . G. C. Todd, Greenfield L. Tomatis, Lyon . B. F. Trump, Baltimore· J. M. Ward, Frederick Officers - ILSI Alex Malaspina, Atlanta - President Peter B. Dews, Boston - Vice President Ulrich Mohr, Hannover - Vice President Roger D. Middlekauff, Washington - Secretary/Treasurer

Respiratory System

  • Upload
    others

  • View
    13

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Respiratory System

Monographs on Pathology of Laboratory Animals

Sponsored by the International Life Sciences Institute

Editorial Board

J. D. Burek, West Point· J. S. Campbell, Ottawa C. C. Capen, Columbus . A. Cardesa, Barcelona RG.Carison, Kalamazoo· D. de Paola, Rio de Janeiro G. Della Porta, Milan . J. L. Emerson, Atlanta F. M. Garner, Rockville . L. Golberg, Research Triangle Park H. C. Grice, N epean . C. C. Harris, Bethesda . R Hess, Basel C. F. Hollander, Rijswijk· G. H. Hottendorf, Syracuse RD. Hunt, Southborough . T. C. Jones, Southborough Y. Konishi, Nara . D. Krewski, Ottawa· R Kroes, Bilthoven H. Luginbuhl, Bern . U. Mohr, Hannover . P. Olsen, Soborg J. A. Popp, Research Triangle Park· J. R Schenken, Omaha R A. Squire, Baltimore· J. Sugar, Budapest S. Takayama, Tokyo . G. C. Todd, Greenfield L. Tomatis, Lyon . B. F. Trump, Baltimore· J. M. Ward, Frederick

Officers - ILSI

Alex Malaspina, Atlanta - President Peter B. Dews, Boston - Vice President Ulrich Mohr, Hannover - Vice President Roger D. Middlekauff, Washington - Secretary/Treasurer

Page 2: Respiratory System

Respiratory System Edited by

T.e.Jones V.Mohr R.D.Hunt

With 279 Figures and 20 Tables

Springer-Verlag Berlin Heidelberg New York Tokyo 1985

Page 3: Respiratory System

Thomas Carlyle Jones, D. V. M., D. Sc. Professor of Comparative Pathology, Emeritus Harvard Medical School New England Regional Primate Research Center One Pine Hill Drive, Southborough, MA 01772, USA

Ulrich Mohr, M. D. Professor of Experimental Pathology Medizinische Hochschule Hannover Institut fur Experimentelle Pathologie Konstanty-Gutschow-Strasse 8 3000 Hannover 61, Federal Republic of Germany

Ronald Duncan Hunt, D. V. M. Professor of Comparative Pathology Harvard Medical School New England Regional Primate Research Center One Pine Hill Drive, Southborough, MA 01772, USA

ISBN-13: 978-3-642-96848-8 e-ISBN-13: 978-3-642-96846-4 DOl: 10.1007/978-3-642-96846-4

Library of Congress Cataloging in Publication Data. Main entry under title: Respiratory system. (Monographs on pathology oflaboratory animals) Bibliography: p. Includes index. 1. Laboratory animals-Diseases. 2. Respiratory organs-Diseases. 3. Ro­dents-Diseases. 4. Rodents as laboratory animals. 5. Pathology, Comparative. I. Jones, Thomas Carlyle. II. Mohr, U. (Ulrich) III. Hunt, Ronald Duncan. IV. Series. SF996.5.R47 1985 599.32'3 84-14048

This work is subject to copyright. All rights are reserved, whether the whole or part of the material is concerned, specifically those of translation, reprinting, re-use of illustrations, broadcasting, reproduction by photocopying machine or similar means, and storage in data banks. Under § 54 of the German Copyright Law where copies are made for other than private use a fee is payable to 'Verwertungsgesellschaft Wort', Munich.

© Springer-Verlag Berlin Heidelberg 1985

The use of registered names, trademarks, etc. in the publication does not imply, even in the absence of a specific statement, that such names are exempt from the relevant protec­tive laws and regulations and therefore free for general use.

Product Liability: The publisher can give no guarantee for information about drug dosage and application thereof contained in this book. In every individual case the respective user must check its accuracy by consulting other pharmaceutical literature.

2123/3140-543210

Page 4: Respiratory System

Foreword

The International Life Sciences Institute (ILSI) was creat­ed to promote cooperative efforts toward solving critical health and safety questions involving foods, drugs, cosmet­ics, chemicals, and other aspects of the environment. The Officers and Trustees believe that questions regarding health and safety are best resolved when government and industry rely on scientific investigations, analyses, and re­views by independent experts. Further, the scientific aspects of an issue should be examined and discussed on an international basis, separate from the political concerns of individual companies. ILSI is pleased to sponsor this set of monographs on the pathology of laboratory animals. This project will be use­ful in improving the scientific basis for the application of pathologic techniques to health and safety evaluation of substances in our environment. The world wide distribu­tion of the authors, editors, and Editorial Board who are creating these monographs strengthens the expectation that international communication and cooperation will al­so be strengthened.

Alex Malaspina President International Life Sciences Institute

Page 5: Respiratory System

Preface

This book on the respiratory system is the second volume of a set pre­pared under the sponsorship of the International Life Sciences Insti­tute (ILSI). One aim of this set on the Pathology of Laboratory Ani­mals is to provide information which will be useful to pathologists, especially those involved in studies on the safety of foods, drugs, chemicals, and other substances in the environment. It is expected that this and future volumes will contribute to better communication on an international basis among people in government, industry, and acade­mia who are involved in the protection of the public health. The arrangement of this volume is based, in part, upon the philosophy that the first step toward understanding a pathologic lesion is its pre­cise and unambiguous identification. The microscopic and ultrastruc­tural features of a lesion that are particularly useful to the pathologist for definitive diagnosis are therefore considered foremost. Diagnostic terms preferred by the author and editors are used as the subject head­ing for each pathologic lesion. Synonyms are listed although most are not preferred and some may have been used erroneously in prior pub­lications. The problems arising in differential diagnosis of similar le­sions are considered in detail. The biologic significance of each patho­logic lesion is considered under such headings as etiology, natural history, pathogenesis, and frequency of occurrence under natural or experimental conditions. Comparison of information available on similar lesions in man and other species is valuable as a means to gain broader understanding of the processes involved. Knowledge of this nature is needed to form a scientific basis for safety evaluations and experimental pathology. Ref­erences to pertinent literature are provided in close juxtaposition to the text in order to support conclusions in the text and lead toward addi­tional information. Illustrations are an especially important means of non verbal communication, especially among pathologists, and there­fore constitute important features of each volume. The subject under each heading is covered in concise terms and is ex­pected to stand alone, but in some instances it is important to refer to other parts of the volume. A comprehensive index is provided to en­hance the use of each volume as a reference. Some omissions are inevitable and we solicit comments from our col­leagues to identify parts which need strengthening or correction. We have endeavored to include important lesions which a pathologist might encounter in studies involving the rat, mouse, or hamster. Newly recognized lesions or better understanding of old ones may make revised editions necessary in the future. The editors wish to express their deep gratitude to all of the individuals who have helped with this enterprise. We are indebted to each author and member of the Editorial Board whose names appear elsewhere in the volume. We are especially grateful to the Officers and Board of Trustees of the International Life Sciences Institute for their support and understanding. Several people have worked directly on important details in this venture. These include Nina Murray, Executive Secre-

Page 6: Respiratory System

VIII Preface

tary; Beverly Blake, Editorial Assistant; June Armstrong, Medical Il­lustrator; and Virginia Werwath, Administrative Assistant. Sharon K. Coleman, ILSI Coordinator for External Affairs, was helpful on many occasions. We are particularly grateful to Dr. Dietrich Gotze and his staff at Springer-Verlag for the quality of the published product.

November 1984 T.C.Jones V.Mohr R.D.Hunt

Page 7: Respiratory System

Table of Contents

The Upper Respiratory System (Nares, Larynx, Trachea) . 1

Histology, Ultrastructure, Embryology . . . . . . . . . . . . 3

Macrosopic, Microscopic, and Ultrastructural Anatomy of the Nasal Cavity, Rat J. A. POPP and N. A. MONTEIRO-RIVIERE ........... 3

Development of Syrian Golden Hamster Tracheal Epithelium During Prenatal and Immediate Postnatal Stages M.EMURA. . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11

Epithelial Alterations in Explant Cultures of Fetal Tracheae of Syrian Golden Hamsters M.EMURA. . . . . . . . . . . . . . . . . . . . . . . . . . . . .. 27

Neoplasms . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33

Response to Carcinogens of Respiratory Epithelium, Syrian Golden Hamster (Mesocricetus Auratus) H.-B. RICHTER-REICHHELM, W. BONING, and J. ALTHOFF. 33

Polypoid Adenoma, Nasal Mucosa, Rat W.D.KERNS .................. .

Neoplasms, Mucosa, Ethmoid Turbinates, Rat S. F. STINSON and H. M. REZNIK-SCHOLLER .

Squamous Cell Carcinoma, Nasal Mucosa, Rat W.D.KERNS .................. .

Squamous Cell Carcinoma, Upper Respiratory Tract, Syrian Hamster

41

47

54

P. M. POUR. . . . . . . . . . . . . . . . . . . . . . . . . . . . 62

Adenocarcinoma, Anterior Nasal Epithelium, Rat S. F. STINSON and G. REZNIK ........... . 67

Hemangiosarcoma, Nasal Cavity, Mouse W. E. GIDDENS Jr. and R. A. RENNE .... 72

Clear Cell Carcinoma, Larynx, Syrian Hamster P.M. POUR .................... . 75

Lesions Due to Infections. . . . . . . . . . . . . . . . . . . . . .. 78

Murine Respiratory Mycoplasmosis, Upper Respiratory Tract, Rat T. R. SCHOEB and J. R. LINDSEY. . . . . . . . . . . . . . . . . 78

Sialodacryoadenitis Virus Infection, Upper Respiratory Tract, Rat D. G. BROWNSTEIN ....................... 84

Page 8: Respiratory System

X Table of Contents

The Lung (Bronchi, Bronchioles, Alveolar Ducts, Alveoli, Pleura) . 87

Histology and Ultrastructure . . . . .

Structure and Function of the Lung C. KUHN III ............ .

Neoplasms.

Bronchiolar/Alveolar Adenoma, Lung, Rat G. A. BOORMAN ............... .

Alveolar Type II Cell Adenoma, Lung, Mouse S. L. KAUFFMAN and T. SATO ........ .

Bronchiolar Adenoma, Lung, Mouse S. L. KAUFFMAN and T.SATO .....

Bronchiolar/Alveolar Carcinoma, Lung, Rat G. A. BOORMAN ................ .

Squamous Cell Carcinoma, Lung, Syrian Hamster P. M. POUR and H. M. REZNIK-SCHULLER.

Squamous Cell Carcinoma, Lung, Rat G. A. BOORMAN ............... .

Radiation-Induced Squamous Cell Carcinoma, Lung of Rodents

89

89

99

99

102

107

112

117

124

F. F. HAHN. . . . . . . . . . . . . . . . . . . . . . . . . . 127

Pleural Mesothelioma, Syrian Hamster A.CARDESAandJ.A.BOMBI ...

Metastatic Tumors, Lung, Mouse B. SASS and A. G. LIEBELT

Nonneoplastic Lesions . . .

Bleomycin-Induced Injury, Mouse: A Model for Pulmonary Fibrosis

133

138

160

D. H. BOWDEN .... . . . . . . . . . . . . . . . . . . 160

Endogenous Lipid Pneumonia in Female B6C3Fl Mice Y. EMI and Y. KONISHI ................. . 166

Pulmonary Lipidosis, Rat Y. EM I, R. HIGASHIGUCHI, and Y. KONISHI . 169

Alveolar Lipoproteinosis, Rat W.WELLER .................. . 171

Bronchiolar/Alveolar Hyperplasia, Lung, Rat G. A. BOORMAN ................ . 177

Fly Ash Pneumoconiosis, Hamster G. E. DAGLE and A. P. WEHNER 180

Asbestosis, Hamster G. E. DAGLE and A. P. WEHNER 183

Pulmonary Hair Embolism A. KAST ............ . 186

Page 9: Respiratory System

Table of Contents XI

Lesions Due to Infection . . . . . . . . . . . . .

Sendai Virus Infection, Lung, Mouse and Rat D. G. BROWNSTEIN ......... .

Rat Coronavirus Infection, Lung, Rat D. G. BROWNSTEIN ......... .

Pneumonia Virus of Mice Infection, Lung, Mouse and Rat

195

195

203

D. G. BROWNSTEIN ................. 206

Sialodacryoadenitits Virus Infection, Lung, Mouse D. G. BROWNSTEIN .............. .

Murine Respiratory Mycoplasmosis, Lung, Rat T. R. SCHOEB and J. R. LINDSEY ........ .

Pneumocystosis, Lung, Rat J. K. FRENKEL ......... .

Aspergillosis and Mucormycosis, Lung, Rat J. K. FRENKEL ............... .

Toxoplasmosis, Lung, Mouse and Hamster J. K. FRENKEL

Subject Index . .

210

213

218

224

227

231

Page 10: Respiratory System

List of Contributors

llirgen Althoff, M. D. Professor of Experimental Pathology, Hannover Medical School, 3000 Hannover 61, Federal Republic of Germany

Josep Antoni Bombi, M. D. Assistant Professor, Department of Pathology, University of Barcelona, Medical School, Barcelona, Spain

W. Boning, Dr. rer. nat. Hannover Medical School, Hannover, Federal Republic of Germany

Gary A. Boorman, D. V. M., Ph. D. Head, Tumor Pathology, Chemical Pathology Branch, NIEHS, Research Triangle Park, North Carolina, USA

Drummond H. Bowden, M. D. Professor and Head, Department of Pathology, University of Manitoba, Manitoba, Canada

David G. Brownstein, D. V. M. Associate Professor of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut, USA

A. Cardesa, M. D. Patologica Facultad de Medicina, Universidad de Barcelona, Barcelona, Spain

Gerald E. Dagle, D. V. M., Ph. D. Staff Pathologist, Battelle, Pacific Northwest Laboratory, Richland, Washington, USA

Yohko Emi, D.V.M. Department of Oncological Pathology, Cancer Center, Nara Medical College, Nara, Japan

Makito Emura, Priv. Doz. Dr. rer. nat. Head, Tissue Culture Unit, Institute of Experimental Pathology, Hannover Medical College, Hannover, Federal Republic of Germany

J. K. Frenkel, M. D., Ph. D. Professor of Pathology and Oncology, Department of Pathology and Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA

W. Ellis Giddens, Jr., D. V. M., Ph. D. Associate Professor, Division of Animal Medicine, Department of Pathology, School of Medicine, University of Washington, Seattle, Washington, USA

Fletcher F. Hahn, D. V. M., Ph. D. Head, Pathology Group, Inhalation Toxicology Research Institute, Lovelace Biomedical and Environmental Research Institute, Albuquerque, New Mexico, USA

Ryuichi Higashiguchi, M. D. Assistant, Department of Oncological Pathology, Cancer Center, Nara Medical College, Nara, Japan

Page 11: Respiratory System

XIV List of Contributors

Alexander Kast, Priv. Doz. Head, Department of Experimental Pathology, Nippon Boehringer Ingelheim Co. Ltd., Hyogo, Japan

Shirley L. Kauffman, M. S., M. D. Professor of Pathology, Department of Pathology, State University of New York, Downstate Medical Center, Brooklyn, New York, USA

William D. Kerns, D. V. M., M. S. Pathologist, Smith Kline & French Laboratories, Philadelphia, Pennsylvania, USA

Yoichi Konishi, M. D. Professor, Department of Oncological Pathology, Cancer Center, Nara Medical College, Nara, Japan

Charles Kuhn, III, M. D. Professor of Pathology, School of Medicine, Washington University, St. Louis, Missouri, USA

Annabel G. Liebelt, Ph. D. Biologist, Registry of Experimental Cancers, National Institutes of Health, Bethesda, Maryland, USA

J. Russell Lindsey, D. V. M., M. S. Professor, Department of Comparative Medicine, Univ. of Alabama, Director, Laboratory Animal Medicine, Veteran's Administration Medical Center, Birmingham, Alabama, USA

Nancy A. Monteiro-Riviere, M.S., Ph.D. Postdoctoral Fellow, Department of Pathology, C. I. I. T., Research Triangle Park, Visiting Assistant Professor, School of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA

James A. Popp, D. V. M., Ph. D. Head, Department of Experimental Pathology and Toxicology, Chemical Industry Institute of Toxicology, Research Triangle Park, North Carolina, USA

Parviz M. Pour, M. D. Professor, Eppley Institute for Research in Cancer, Department of Pathology and Laboratory Medicine, University of Nebraska Medical Center, Omaha, Nebraska, USA

Roger A. Renne, D. V. M. Biology and Chemistry Department, Battelle, Pacific Northwest Laboratory, Richland, Washington, USA

Gerd Reznik, D. V. M., Priv. Doz. Pathology Services Project, National Center for Toxicological Research, Jefferson, Arkansas, USA

Hildegard M. Reznik-Schuller, D. V. M., Priv. Doz. Associate Professor of Experimental Oncology, Acting Chief, Laboratories of Experimental Therapeutics and Metabolism, NCI, Division of Cancer Treatment, Bethesda, Maryland, USA

H. B. Richter-Reichhelm, D. V. M. Priv. Doz. for Experimental Pathology, Max von Pettenkofer Institut, Bundesgesundheitsamt, Berlin, Federal Republic of Germany

Bernard Sass, D. V. M., M. S. Senior Investigator, Registry of Experimental Cancers, National Institutes of Health, Bethesda, Maryland, USA

Page 12: Respiratory System

List of Contributors XV

Tamiko Sato, M. D. Associate Professor of Anatomy, Department of Anatomy, New York Medical College, Valhalla, New York, USA

Trenton R. Schoeb, D. V. M., Ph. D. Assistant Professor of Comparative Medicine, Schools of Medicine and Dentistry, University of Alabama, Birmingham, Alabama, USA

Sherman F. Stinson, Ph. D. Frederick Cancer Research Center, Frederick, Maryland, USA

Alfrj;)d P. Wehner, D. M. D., D. D. S., Sc. D, cando med. Task Leader, Industrial Toxicology, Battelle, Pacific Northwest Laboratory, Richland, Washington, USA

W. Weller D. V. M. Silikose-Forschungsinstitut der Bergbau-Berufsgenossenschaft, Bochum, Federal Republic of Germany

Page 13: Respiratory System

The Upper Respiratory System (Nares, Larynx, Trachea)

Page 14: Respiratory System

HISTOLOGY, ULTRASTRUCTURE, EMBRYOLOGY

Macroscopic, Microscopic, and Ultrastructural Anatomy of the Nasal Cavity, Rat

James A. Popp and Nancy A. Monteiro-Riviere

For those interested in experimental studies of the nasal cavity, it is important first to understand the normal structure. This includes macroscopic, mi­croscopic, and ultrastructural anatomic character­istics of the nasal cavity, surface epithelium, and submucosa. The medial surface of the three major turbinates is exposed when a midsagittal cut of a rat's nose is made and the septum is removed (Fig. 1). The na­soturbinate is located on the dorsal and anterior part of the nasal cavity, while the maxilloturbinate is located on the ventral and anterior part. The na­soturbinates and maxilloturbinates have relatively flat medial surfaces. The ethmoid turbinates con­sist of several lamellae : dorsal and ventral lamella of endoturbinate II, endoturbinate III, and a dorsal and ventral lamella of endoturbinate IV (Hebel and Stromberg 1976). Each endoturbinate has a flat medial surface and the more dorsal tur­binate is larger than the ventral endoturbinates. Complete histological evaluation of normal turbi­nates requires examination of multiple levels to determine the distribution of epithelial types and submucosal glands and to determine variations within a single epithelial type based on location in the nasal cavity. Multiple sections are also re-

ABC 0

Fig.t. Schematic illustrations of the rat nasal cavity. Left: inverted rat skull with palatine structures as reference points for making cross sections ofthe nose. Right: sagittal

quired to identify and characterize lesions which may be localized in a small part of the nasal tis­sue. To achieve a uniform histological examina­tion of the nasal passages, several groups have de­veloped rather precise methods for making cross sections of the nose (Young 1981; Chang et al. 1983). One such uniform method is demonstrated in Fig.1. The blocks of tissue are hand cut with the previously decalcified skull inverted, and the location of the cross sections is determined by palatine structures. The first cut is made just ante­rior to the incisor teeth. The second cut is made halfway between the base of the incisor teeth and the incisive papilla. The third cut is made directly through the incisive papilla, while the fourth cut is made over the second palatal ridge, and the fifth cut is made through the second molar teeth. The resulting four blocks of tissue are embedded in ei­ther paraffin or glycol methacrylate with the ante­rior face down. Although this procedure provides a uniform sampling of the nasal structures, some alteration in the location of these sections may be necessary in specific experimental studies. The structures of the various nasal cavity surfaces at the locations defined above are indicated in Fig.2. At level A the nasoturbinate is attached to

section. Nasoturbinate (n), maxilloturbinate (m), and eth­moid turbinates (e). Lines indicate the location of sections taken for light microscopic examination

Page 15: Respiratory System

4 James A. Popp and Nancy A. Monteiro-Riviere

the dorsal lateral wall and extends a shorter dis­tance into the nasal cavity in contrast to the sec­tion at level B. The maxilloturbinate is also less extensive and is attached to the ventral lateral wall of the nasal cavity. At level B, the nasoturbi­nate extends from the dorsal wall and projects ventrally to half the depth of the nasal cavity. Note that this turbinate turns laterally and dorsal­ly, producing a hook in the cross section of turbi­nate. The maxilloturbinate is attached to the lat­eral wall and projects dorsally into the nasal cavity. The nasolacrimal duct lies ventral to it. In this section one can see the vomeronasal organ lo­cated in the ventral portion of the nasal septum. Although the function of this organ is still under investigation, it has a sensory function and is in­volved in pheromone-mediated behavior (Vac­carezza et al. 1981). At level C, the ethmoid turbi­nate appears to be free in the nasal cavity, since only the tip of the dorsal endoturbinate is in­cluded at this level. The nasoturbinates and max-

Fig. 2. A Transverse section through A, (Fig. 1) x 16; B transverse section through B, x 9; C transverse section through C, x 9; D transverse section through D, x 9. n, nasoturbinate; m, maxilloturbinate; e, ethmoid turbinate; s, nasal septum; w, lateral wall; v, vomeronasal organ; 0, location of septal olfactory organ; p, nasopharynx; d, nasolacrimal duct

illoturbinates are not present at this level. Since the palatine landmark for this section is through the incisive papilla, this structure is frequently ob­served on the palatine surface of the section. If the section is through the small nasal palatine ducts, stratified squamous epithelium is observed lining the ducts at the point where they connect the na­sal and oral cavities. The fourth section (level D) is through the center of the ethmoid turbinate, which forms a complicated set of lamellae (scrolls) arising from the dorsal and lateral nasal walls. The nasopharynx is ventral to the! ethmoid turbinates. In specific virus-free rats, as defined by a standard rat murine viral antibody screening procedure (Microbiological Associates, Bethesda, Maryland), small lymphocyte accumulations are routinely found adjacent to this level of the naso­pharynx, while leukocytes are not observed at other locations in the rat nasal mucosa. This small bit of lymphoid tissue adjacent to the naso­pharynx is also seen consistently in mice.

Page 16: Respiratory System

Macroscopic, Microscopic, and Ultrastructural Anatomy of the Nasal Cavity, Rat 5

The nasal cavity is lined by three types of epithe­lium: squamous, respiratory, and olfactory. Squa­mous epithelium covers the nasal vestibule and the anterior tip of the nasoturbinate and maxillo­turbinate and extends posteriorly as a narrow zone along the ventral nasal surface to the nasal palatine ducts. Respiratory epithelium covers all of the maxilloturbinate and most of the nasoturbi­nate except for its dorsal attachment, and also ex­tends onto the anterior and ventral parts of the ethmoid turbinates. Olfactory epithelium covers the ethmoid turbinates, but also extends along the dorsal wall of the anterior nasal cavity to include the attachment of the nasoturbinate. The nasal septum is covered by respiratory epithelium ex­cept for some squamous epithelium in the area of the vestibule and olfactory epithelium on the dor­sal attachment. A small oval area of olfactory epithelium exists on the ventral nasal septum just anterior to the septal window and is not contiguous with other olfacto­ry epithelium. This focal area of olfactory epithe­lium is frequently referred to as the septal olfacto­ry organ or the organ of Rodolfo-Masera, and may function as a detection mechanism during quiet respiration (Rodolfo-Masera 1943; Adams and McFarland 1971). The zones of demarcation between any two of the epithelial types are very abrupt, as is evident by either light or electron microscopy. Using morphometric procedures, the volume, to­tal surface area, and surface area lined by each epithelial type have been quantitated for the nasal cavities of both rats and mice (Gross et al. 1982). In 16-week-old male Fischer-344rats, the nasal cavity has a volume of approximately 250 mm3

and a surface area of approximately 1350 mm2• In 16-week-old male B6C3Fl mice, the nasal volume is approximately 32 mm3 and the surface area is approximately 290 mm2• This large surface area is important in the warming, cleansing, and humidi­fication of inspired air. Squamous epithelium covers 3% of the surface area in rats and 7% in mice, while the remainder of the surface is equally covered by respiratory (47% rats; 46% mice) and olfactory epithelium (50% rats, 47% mice). Detailed light microscopy of the surface epitheli­um of the nasal cavity has been completed. While no unique or surprising characteristics of the squamous epithelium have been described, inter­esting observations of the respiratory epithelium have been made. The nasal respiratory epithelium has been generally described as pseudostratified ciliated columnar epithelium. While this histolog­ical description is correct for the respiratory epi-

thelium found in some areas of the nasal cavity, other areas do not fit this general description. The respiratory epithelium in some areas, particu­larly the more anterior segments of the maxillo­turbinates and nasoturbinates, consists of either cuboidal or nonciliated columnar cells, which may be found either alone or interspersed with a few ciliated cells (Fig. 3). Goblet cells are scattered unevenly throughout the respiratory epithelium and are most numerous in the nasal septum. In general, goblet cells are also relatively numerous in the ventral respiratory epithelium, particularly at the junction with squamous epithelium. Histo­logically, olfactory epithelium has a uniform pseudostratified columnar structure (Fig. 4). It is composed primarily of olfactory cells (bipolar neurons) and sustentacular cells, although a sin­gle row of basal cells is found adjacent to the bas­allamina. The intertwined cells make it impossi­ble to distinguish individual cell borders. Nuclei are approximately six deep and covered with a nuclear-free zone of cytoplasm at the apical end. A thin eosinophilic zone composed of cilia and olfactory vesicles is present adjacent to the nasal cavity. A thin mucous layer is found on the surface of the olfactory and respiratory epithelium. The compo­sition and function of this mucous blanket has been recently reviewed (Widdicomb and Wells 1982; Proctor 1982). The mucous layer consists of a superficial layer of mucus and an underlying watery periciliary fluid. The continuously moving layer is the first defense of the nasal cavity against inhaled gases and particles. Mucus is continually produced, flows on the nasal surfaces due to ciliary acitivity, and is ultimately swallowed after passing through the nasopharynx. The submucosal zone of the nasal passages is extremely vascular, although the vascularity is greatest in the nasoturbinates and maxilloturbi­nates. Between the numerous and relatively large vessels, 15-20 glands have been described in the submucosa of the septum, lateral wall, nasoturbi­nate, maxilloturbinate, and ethmoid turbinate (Bojsen-Moller 1964). The glands underlying re­spiratory epithelium are both serum and tp.ucus producing, with individual clusters of glandular tissue connected by ducts which pass anteriorly. Ducts of the serous glands reportedly empty into the vestibule of the nasal cavity (Bojsen-Moller 1964). The ducts open on inspiration and close on expiration, thereby releasing the glandular con­tent of the serous glands only to incoming air to aid in the humidification of the air. Ducts of the mucous glands empty into the vomeronasal or-

Page 17: Respiratory System

6 James A. Popp and Nancy A. Monteiro-Riviere

Fig. 3 (Above). Light micrograph of respiratory epithelium. Cilliated (c) and non ciliated columnar (arrowhead) cells. Note the glands (g), duct (d), and blood vessel (b) in the submucosa. x 480

gan. In contrast to the different types of glands under the respiratory epithelium, the olfactory re­gion has only a simple tubular mucus-producing gland (Bowman's gland), which opens directly on the surface (Bojsen-Meller 1964). Ultrastructural studies of toxin-induced lesions in the nasal mucosa have been published previous­ly; however, the ultrastructural characteristics of the normal nasal structures had been incomplete­ly described until recently (Monteiro-Riviere and Popp 1984). Transmission electron microscopic (TEM) studies of the respiratory epithelium dem-

Fig.4 (Below). Transition between olfactory (arrowhead) and respiratory epithelia. Bipolar neurons (n) and basal (b) and sustentacular (s) cells can be seen in the olfactory part, while ciliated (c) and goblet (g) cells can be seen in the re­spiratory portion. x 640

onstrated six distinct cell types: basal, cuboidal, nonciliated columnar, ciliated, brush, and goblet cells. The ciliated, basal, and goblet cells are simi­lar to the comparable cell types described in other locations within the respiratory system. The cu­boidal cell has sparse microvilli but no other dis­tinctive ultrastructural characteristics (Fig. 5). The nonciliated columnar cell has an extensive accu­mulation of smooth endoplasmic reticulum in the apical cytoplasm (Fig. 6). The accumulation of this organelle suggests that nonciliated columnar cells may be the source of cytochrome P 450 and

Page 18: Respiratory System

Macroscopic, Microscopic, and Ultrastructural Anatomy of the Nasal Cavity, Rat 7

Fig.S (Above). A brush cell (B) and cuboidal cells (C) in re­spiratory epithelium lining the nasal cavity. Microvilli of brush cell protruding above (arrow) adjacent cuboidal cells. TEM, x 7400

Fig. 6 (Below). Two nonciliated columnar, cells (C) in respi­ratory epithelium. Microvilli (M) and extensive smooth en­doplasmic reticulum (arrow) can bee seen in the apical re­gion of the cell. TEM, x 14400 (Monteiro-Riviere and Popp 1984)

Page 19: Respiratory System

8 James A. Popp and Nancy A. Monteiro-Riviere

P450-associated enzymes that have been previ­ously described in the nasal mucosa (Hadley and Dahl 1982). Unfortunately, information is not yet available on specific cell localization of P450 in the respiratory epithelium. The brush cell, with distinctive ultrastructural characteristics, has re­cently been described in the rat nasal respiratory epithelium (Monteiro-Riviere and Popp 1984). It is pear-shaped with a large basal part containing the nucleus, while the narrow apical surface ex­tends into the nasal cavity (Fig. 5). Nonbranching microvilli cover the small surface. These microvil­li are much longer and wider than microvilli of adjacent nonciliated cells, but are shorter than cilia. The apical cytoplasm has bundles of fila­ments and numerous clear vesicles. Paired cis­ternae are frequently seen in the supranuclear region. Although it has been hypothesized that brush cells in other locations may function as chemoreceptors, baroreceptors, or stretchrecep­tors (Meyrick and Reid 1968; Luciano et al. 1968, 1981), the function of this cell in the nasal cavity is unknown. In the rat nasal respiratory epithelium, intraepi­thelial nerve endings containing both clear and dense vesicles have been observed most frequent-

ly adjacent to the basal lamina (Fig. 7) (Monteiro­Riviere and Popp 1984). They do not have a pref­erentiallocation adjacent to any specific cell type. The nerve endings and nerves in the respiratory epithelium and submucosa are branches of the tri­geminal nerve and have a sensory function (Boj­sen-M011er 1975). When examined by TEM, olfactory epithelium consists of three distinct cell types: sustentacular (supporting), olfactory (bipolar neuron), and bas­al cells (Frisch 1967). The bipolar neuron has an apical olfactory vesicle from which immotile cilia project in all directions. The sustentacular cell has long microvilli on the apical surface and pigment granules in the cytoplasm which account for the brown color of the olfactory epithelium noted up­on gross observation. Scanning electron microscopy (SEM) of the nor­mal nasal cavity clearly demonstrates that much of the surface is covered by a relatively smooth layer of mucus. When the mucous layer is re­moved, SEM allows one to study the surface char­acteristics of cells and determine the distribution of the various cell types on the basis of their sur­face structure. SEM dramatically demonstrates the uneven distribution of ciliated cells in the re-

Fig. 7. An intraepithelial axon (A) located just above basal lamina (BL) in respiratory epithelium. Clear vesicles (arrow), mitochondria, and neurotubules are present. TEM, x 33000 (Monteiro-Riviere and Popp 1984)

Page 20: Respiratory System

Macroscopic, Microscopic, and Ultrastructural Anatomy of the Nasal Cacity, Rat 9

Fig.8 (Above). Ciliated (C), nonciliated columnar (NC), and brush cell (arrowhead) in the respiratory epithelium. SEM, x 1800

Fig.9 (Below). Olfactory epithelium. Note the tangled web of cilia (arrowhead). SEM, x 4500

Page 21: Respiratory System

10 James A. Popp and Nancy A. Monteiro-Riviere

spiratory epithelium (J. A. Popp and J. T. Martin, unpublished work) (Fig. 8). In general, the anteri­or respiratory epithelium is nonciliated on the na­soturbinate, maxilloturbinate, and lateral wall. The surfaces become progressively more ciliated from anterior to posterior. However, the various surfaces are not equally ciliated at a single cross­sectional level of the nasal cavity. For example, in the middle third of the nasoturbinate and maxillo­turbinate, approximately 70% of the medial sur­face of the nasoturbinate is covered with ciliated cells while only 15% of the medial surface of the maxilloturbinate is covered with ciliated cells. In nonciliated areas, brush cells are easily identified due to their small surface area and long apical microvilli. They compose less than 1 % of the sur­face area, accounting for their infrequent observa­tion in TEM studies. In contrast to the variable surface features in re­spiratory epithelium, SEM reveals the olfactory epithelium to be relatively uniform. The surface of the tissue is covered by a tangled web of cilia, although the tip of the olfactory vesicles may occasionally be observed in some locations (Fig. 9). A thorough understanding of the macroscopic, microscopic, and ultrastructural characteristics of the normal rodent nasal cavity has lagged behind experimental studies with nasal toxicants. A prop­er interpretation of lesions in the rat nasal cavity can be achieved only when one has a basic under­standing of the normal nasal passages.

References

Adams DR, McFarland LZ (1971) Septal olfactory organ in Peromyscus. Comp Biochem Physiol (A) 40: 971-974

Bojsen-Moller F (1964) Topography of the nasal glands in rats and some other mammals. Anat Rec 150: 11-24

Bojsen-Moller F (1975) Demonstration of terminalis, ol­factory, trigeminal and perivascular nerves in the rat na­sal septum. J Comp Neuro1159: 245-256

Chang JC, Gross EA, Swenberg JA, Barrow CS (1983) Na­sal cavity deposition, histopathology and cell prolifera­tion after single or repeated formaldehyde exposures in B6C3F1 mice and F344 rats. Toxicol Appl Pharmacol 68: 161-176

Frisch 0 (1967) Ultrastructure of mouse olfactory mucosa. AmJ Anat 121: 87-120

Gross EA, Swenberg JA, Fields S, Popp JA (1982) Com­parative morphometry of the nasal cavity in rats and mice. J Anat 135: 83-88

Hadley WM, Dahl AR (1982) Cytochrome P-450 depen­dent monooxygenase activity in rat nasal epithelial membranes. Toxicol Lett 10: 417-422

Hebel R, Stromberg MW (1976) Anatomy of the laborato­ry rat. Williams and Wilkins, Baltimore

Luciano L, Reale E, Ruska H (1968) Ueber eine 'chemo­rezeptive' Sinneszelle in der Trachea der Ratte. Z Zell­forsch 85: 350-375

Luciano L, Castellucci M, Reale E (1981) The brush cells of the common bile duct of the rat. This section, freeze­fracture and scanning electron microscopy. Cell Tissue Res 218: 403-420

Meyrick B, Reid L (1968) The alveolar brush cell in rat lung - a third pneumonocyte. J Ultrastruct Res 23: 71-80

Monteiro-Riviere NA, Popp JA (1984) Ultrastructural characterization of the nasal respiratory epithelium in the rat. Am J Anat 169: 31-43

Popp JA, Martin JT (1984) Surface topography and distri­bution of cell types in the rat nasal respiratory epitheli­um: scanning electron microscopic observations. Am J Anat (in press)

Proctor OF (1982) The mucociliary system. In: Proctor OF, Andersen I (eds) The nose: upper airway physiolo­gy and atmospheric environment. Elsevier, New York, p245-278

Rodolfo-Masera DT (1943) Sui'esistenza di un particolare organo olfattivo nel sette nasale della cavia e di altri roditori. Arch Ital Anat Embriol48: 157-212

Vaccarezza OL, Sepich LN, Tramezzani JH (1981) The vomeronasal organ of the rat. J Anat 132: 167-185

Widdicomb JG, Wells UM (1982) Airway secretions. In: Proctor OF, Andersen I (eds) The nose: upper airway physiology and atmospheric environment. Elsevier, New York, p 215-224

Young JT (1981) Histopathologic examination of the rat nasal cavity. Fund Appl Toxicoll: 309-312

Page 22: Respiratory System

Development of Syrian Golden Hamster Tracheal Epithelium 11

Development of Syrian Golden Hamster Tracheal Epithelium During Prenatal and Immediate Postnatal Stages

Makito Emura

The Syrian golden hamster (Mesocricetus auratus) makes an excellent model for studies on chemical carcinogenesis of the respiratory tract, and in par­ticular the trachea (Wynder and Hecht 1976). This organ is particularly sensitive to N-nitroso com­pounds, among others. It is also possible, using these compounds, to induce tumors transplacen­tally (Mohr 1973). For the study of the so-called early changes in animals exposed to strong chemi­cal carcinogens, an understanding of the develop­ment of the trachea is necessary (Mohr et al. 1979). The anlage of the trachea in the fetus can­not be easily distinguished until after the 9th day of pregnancy, but only a few days later signs of rapid growth and differentiation are clearly recog­nizable.

Predifferentiation Stage

Light Microscopy. The tracheobronchial rudi­ments of the Syrian hamster become independent of the early esophageal ducts (Fig. 10) between the 9th and 10th gestational days. The next stage, ex­tending from the 10th to 11th gestational days, can be regarded as the predifferentiation stage, since no marked sign of differentiation is detected

Fig. to (Left). Longitudinal sections of tracheal epithelium, Syrian hamster on the 10th gestational day. Trachea (T) and esophagus (OE) with ventral (top) and dorsal epitheli­um (bottom). H, heart. Hand E, x 43

in the epithelium either by light or electron mi­croscopy. At this stage the tracheal epithelium is mainly composed of one layer of tall and narrow columnar cells (Fig. 11); the nuclei are elongated, ovoid, or round and basally situated. At the lumi­nal surfaces, the epithelium also contains a few el­lipsoid or polygonal cells. Several cells possess cy­toplasmic vacuoles. In a relatively few epithelial cells, the luminal portion of the cytoplasm is PAS positive. When pretreated with diastase, very few cells subsequently react to PAS and none stain with alcian blue.

Electron Microscopy. At this stage no signs of dif­ferentiation can be detected in the cells and no particular cell types are discernible (Fig. 12). The irregular luminal surface usually possesses sparse cytoplasmic projections or microvilli of various lengths (Figs. 13 and 14). The nuclei are round to ovoid and their contours are mostly smooth and usually contain two to four nucleoli. The nuclear chromatin fibrils are uniformly dispersed in the nucleoplasm of epithelial cells and none of the chromatin condensations of the type seen in ma­turing cell types are recognizable until the 12th day. In the stromal fibroblasts, however, such chromatin condensations are already beginning

Fig.11 (Right). Trachea at higher magnification. Hand E, x 106

Page 23: Respiratory System

12 Makito Emura

Fig.12 (Upper left). Epithelial cells on the 11th gestational day. Note sparse endoplasmic reticulum (ER), abundance of free polyribosomes and glycogen granules (G), uni­formly diffuse nuclear chromatin fibrils, smooth contours of nuclei, distinct nucleolonemas of the nucleoli (NL), and intercellular spaces. In comparison with the 10th gestation­al day, the only difference is the absence of cytoplasmic vacuoles and vesicles. Centrioles (C) and small cytoplas­mic projections can be seen. TEM, uranyl acetate and lead citrate, x 5070

Fig.13 (Lower left). Epithelial cell on the 11th gestational day. Note the small amount of ER and glycogen (G). TEM, x 11640

Fig. 14 (Upper right). a Vesicles in the epithelial cells on the 10th gestational day. Note relatively large polyribosomes in the cytoplasm and vesicles (arrow), which begin to re­semble ER. TEM, x 19890. b Accumulation of glycogen (G) can be observed around the protrusions and vesicle membrane. Arrow indicates ribosomes attached to mem­brane. TEM, uranylacetate and lead citrate, x 11230

Fig. 15 (Lower right). A solitary immature cilium projecting from the luminal surface. TEM, uranyl acetate and lead citrate, x 11640

Page 24: Respiratory System

Development of Syrian Golden Hamster Tracheal Epithelium 13

to occur. Glycogen granules are either scattered throughout the cytoplasm or accumulate in a small part of the cytoplasm (Fig. 13). Some cells possess one or two cilia (Fig. 15). In the luminal part of the cytoplasm a few centrioles can occa­sionally be observed (Figs.12 and 15) and pe­culiar tightly bound intercellular junctions are formed directly against the lumen (Figs.12 and 13). The mitochondria are round to elongated, frequently club-shaped, and their matrices con­tain dense or sparse fine fibrillar or granular ma­terials. Free polyribosomes prevail. Distinct but not well-developed Golgi apparatus assume mainly lamellar structures with some vesi­cles, mostly located near the nuclei (Fig. 13). The basement membrane is distinct and further depo­sition of fibrillar material progresses. On the 10th gestational day, the smooth and rough endoplasmic reticulum (ER) of the epithe­lial cells seem poorly developed and in most cases flattened sac or saccule forms are found. The out­er nuclear membrane in these cells very frequently has widely distributed extranuclear protrusions (Emura 1978) and often contains membranous structures. On the 11th gestational day, the smooth and rough ER increases only slightly (Fig. 13) and the frequency and size of the protrusions extending from the outer nuclear membrane decrease re­markably. The rough- and smooth-membrane­bound vacuoles and vesicles also notably de-

Fig. 16. Longitudinal section of tracheal epithelium. Cranial ventral section. A cartilaginous mesenchyme condensate (M), cells with hemispherical luminal apices and basophilic cytoplasm (black arrows), and basally situated cells (white arrows) are shown. Hand E, x 170

Fig.17. Longitudinal section of tracheal epithelium. Caudal, dorsal section. Note cell with flat luminal surface and eosinophilic cytoplasm (thick arrow). Hand E, x 170

crease in number. However, around the periphery of the nuclei, peculiar intranuclear membranous inclusions appear at intervals and their frequency increases as time progresses. The possibility that these vacuoles and vesicles contribute to the formation of ER cannot be excluded, since in vertebrate and invertebrate oocytes (Wischnitzer 1974) and in embryonic epi­thelium of chick choroidal plexus (Birge and Doolin 1974) the rough ER has been demon­strated to originate in vesicles derived from the outer nuclear membrane.

Early Morphological Indication of Differentiation

Light Microscopy. On the 12th gestational day, the epithelial cells are somewhat flatter than those seen on previous days. No distinct cell types are detected by light microscopy. On the 13th gesta­tional day, approximately 20 horseshoe-shaped cartilaginous condensates consisting of mesen­chymal cells are formed in the trachea (Fig.16). In the epithelium, three cell types can be dis­tinguished. These are tentatively designated as type I, type II, and type III cells in this report. The type I cells feature a hemispherical luminal apex protruding into the lumen, and are charac­terized by a somewhat basophilic or less eosino­philic cytoplasm (Fig.16). Flat luminal surfaces and somewhat eosinophilic or less basophilic

Page 25: Respiratory System

14 Makito Emura

cytoplasm characterize the type II cells (Fig. 17). Cells of this type are most prominent in the dorsal epithelium and are not positive to PAS. The type III cells are basally situated and have oblong, triangular, or polygonal shapes (Fig. 16). The pres­ence of type I and type III cells causes the epithe­lium to assume a double-layered appearance in

Fig.1S (Above). Luminal cytoplasm of cells with hemis­pherical surfaces (type I) and cells with flat luminal sur­faces (type II). Note the abundance of rough ER. Dictyo­some (DCT). TEM, uranyl acetate and lead citrate, x 11640

Fig.19 (Lower left). Luminal part of a type II cell on the 13th gestational day. Note fibrillar material interspersed with dark granules, 21 x 21 to 87 x 114 nm in size. TEM, x 22770

parts, i. e., a luminal and basal layer. Several ep­ithelial cells react positively to PAS, but after pre­treatment with diastase practically no cell has a positive reaction to PAS.

Electron Microscopy. On the 12th gestational day, the luminal apices in most epithelial cells pro-

Fig. 20 (Lower right). Luminal part of a cell resembling type I, on the 13th gestational day. Fibrillar material inter­spersed with dark granules can be seen (26 x 53 to 42 x 65 nm). Note the proximity of the fibrillar material to existing centriole. TEM, uranyl acetate and lead citrate, x 22770

Page 26: Respiratory System

Development of Syrian Golden Hamster Tracheal Epithelium 15

trude slightly into the lumen, although their lumi­nal surfaces are still irregular. The scattered gly­cogen granules have diminished in many of the cells; the ER is not well developed, possesses rough surfaces, and assumes flattened sac-like forms. The outer nuclear membrane again starts to form circumscribed extensions, which are small but similar to those observed on the 10th gesta­tional day. Membranous and vesicular intranu­clear inclusions at the nuclear periphery occur more frequently than on the 11 th gestational day. The smooth- and rough-membrane-bound vacuoles and vesicles already seen on the 10th gestational day occur again, although only occa­sionally. The nuclei are still round to ovoid in shape with smooth contours. Distinct condensations of nu­clear chromatin fibrils occur in 20%-30% of ep­ithelial cells along the nuclear envelope, as well as in the inner area of the nuclei, although this is much less extensive than in maturing fetal mu­cous cells. On the 13th gestational day, three cell types can be distinguished in the epithelium. The first type

Fig.21 (Upper left). Longitudinal sections of the ventral epithelium (pars cartilaginea). No ciliated cel\s are seen. Cytoplasmic vesicles are prominent. Hand E, x 170

Fig. 22 (Lower left). Longitudinal sections of dorsal epithe­lium (pars membranacea). A few obviously ciliated cel\s

(I) is composed of cells with smooth hemispheri­cal luminal surfaces protruding into the lumen (Fig. 18). The second type (II) consists of cells with flat luminal surfaces, on which several short microvilli or cytoplasmic projections can be ob­served (Figs. 18 and 19). The ventral and lateral epithelial cells are largely composed of type I cells. In the dorsal epithelium, type II cells seldom occur; type I cells still predominate. Cells of both types often contain one or two regional accumula­tions of a considerable amount of vesicular and tubular smooth ER in the luminal apices (Figs. 18 and 20). With these accumulations of smooth ER, frequently found in both cell types, dictyosomes develop which are composed of three to seven cis­ternae (Figs. 18 and 20). In the luminal cytoplasm, the rough ER and free polyribosomes are frequently more abundant in the type I than in the type II cells. However, the basal cytoplasm of both cell types apparently con­tains the same amount of rough ER. In a small number of type I cells, fairly numerous ER vesi­cles surrounded by a partly rough membrane can be observed. Generally, in both types of cells, rough ER increases only fractionally in compari-

(C), type II cel\s (II) , and type III cel\s (III) classified on the 13th gestational day. Hand E, x 170

Fig.23 (Right). Longitudinal section of cranial part of the ventral tracheal epithelium on the 16th gestational day. Note ciliated cel\ (arrow). Hand E, x 170

Page 27: Respiratory System

16 Makito Emura

son with previous days of development. Connec­tion of smooth ER with Golgi (or dictyosome) cis­ternae is occasionally encountered. Located near the smooth ER accumulations and dictyosomes sometimes found in type II cells are small areas of fine fibrillar material, often inter­spersed with a few dark granules, ranging from 21 x21 to 87-114nm (average 49x65nm) in size. These are identified as structures similar to "proliferative elements" (Dirksen and Crocker 1966) (Figs. 19 and 20), and they also occur in the luminal cytoplasm of type I cells, although much less frequently. The inner sections of this fibrillar material are usually devoid of ribosomes. The third type (III) of cells are basally situated in the epithelium and resemble type I cells, except that they have no free luminal surfaces. In cells of all three types, as well as the stromal

fibroblasts, a few immature-looking cilia and cen­trioles occasionally occur. On the 13th gestational day, the circumscribed ex­tensions of outer nuclear membrane in the tra­chea, also encountered in the three epithelial cell types and in stromal fibroblasts, are more fre­quent and more conspicuous than on the previous day. Their frequency and size are similar to those of the 10th gestational day. The vesicular and tubular intranuclear inclusions are also more frequent and more conspicuous than on the 11th and 12th days. However, the rough- and smooth-membrane-bound vacuoles and vesicles, which persistently occur, are not so frequent as in the differentiating type II cells on the 14th gestational day. These vacuoles and vesi­cles appear to fuse occasionally with the preexist­ing rough ER.

ig.24 (Above). Type II (ciliated) cell on the 14th gestation­al day. ote ab ence of rough ER. Uranyl acetate and lead citrate x 16640

Fig.25 (Below). Higher magnification of Fig. 24. Procentri­oles (PC): "conden ation form" (CF). Uranyl acetate and lead citrate. x 31590

Page 28: Respiratory System

Development of Syrian Golden Hamster Tracheal Epithelium 17

Golgi cisternae are sometimes connected to the nuclear envelope. Condensation of nuclear chro­matin occurs more extensively on the 13th gesta­tional day in 80%-95% of cells of all types, in­cluding the stromal fibroblast.

Differentiation of Ciliated Cells

Electron microscopy reveals that the primary stage of ciliogenesis takes place on the 13th gesta­tional day in some of the type II cells. However, the first ciliated cells can be detected on the 14th gestational day in the dorsal epithelium (pars membranacea) by both light and electron micros­copy.

Light Microscopy. The epithelium on the 14th ges­tational and following days of development also has a double-layered appearance. The ciliated cells possess granular eosinophilic cytoplasm and flat luminal surfaces, which are features similar to those of the type II cells of the 13th gestational day (Figs.21 and 22). At the caudal part of the dorsal epithelium these cells are usually cylindri­calor cuboidal, sometimes oblong, and extend from the basement membrane to the lumen. In the cranial part of the epithelium, they often assume either conchoidal or bell shapes. Ciliated cells are sparse in the ventral and lateral epithelium (pars cartilaginea). In most of the trachea, the cilia are often longer in the cranial part than in the caudal part of the epithelium. The type II cells observed on the 13th gestational day (Fig.17) are still encountered frequently on the 14th gestational day, particularly in the middle to caudal part of the dorsal epithelium. On the 15th and 16th gestational days, ciliated cells are only found sporadically in the ventral and lateral epithelium (Fig.23). On the last gestational day, the cells are well developed in the dorsal epitheli­um but occur less frequently in the ventral and lat­eral epithelium.

Electron Microscopy. On the 14th gestational day, the three cell types distinguished on the previous day develop more distinct features. On this day the type II cells are at various stages of organellic differentiation. They possess flat luminal surfaces and occur more frequently in the dorsal epitheli­um than on the previous day. A small amount of smooth ER and a few ribosomes exist; these can occasionally be seen in cells of the same type on the previous day. Some cells are seen in various stages of ciliogenesis (Figs. 24-31).

In cells at an early stage of ciliogenesis, the fibril­lar material alone or with the dark granules, 39 x 42 to 73 x 96 nm (average 53 x 66 nm), occurs more frequently than on the 13th day. Among these granules much darker bodies, 63-148 nm in diameter, or larger hollow bodies, 148-208 nm and 63 -1 04 nm in outer and inner diameters re­spectively, are often identified as "condensation forms" (Dirksen and Crocker 1966) (Figs. 24-29). Tubular and vesicular structures are often located near areas of dark granules (Figs.27-30). In cells at a different stage of ciliogenesis, several pro­centrioles, measuring between 116 x 127 and 158 x 180 nm, are associated with the condensa­tion forms. In more advanced cells, almost com­plete centrioles measuring 180-380 nm and small­er, denser condensation forms 48-95 nm in di­ameter occur (Fig. 29). These centrioles, which eventually become ciliary basal bodies, are formed by a process which Anderson and Bren­ner (1971) termed "acentriolar basal body forma­tion," in which the centrioles develop together with structures which bear no resemblance to cen­trioles. The process of ciliogenesis described here seems to correspond to this theory. In developing fetal rats (Stockinger and Cireli 1965; Dirksen and Crocker 1966) and mice (Frisch and Farbman 1968) such fibrogranular material has been re­ported. Stockinger and Cireli (1965) suggested that this fibrogranular material was formed de novo without any influence of preexisting mature centrioles and that the granular materials which were considered to be precentrioles developed through various intermediate stages into mature centrioles. Dirksen and Crocker (1966) found a direct link between mature centrioles and this fi­brogranular material and termed them "prolifera­tive elements." In the fetal rat, Sorokin (1968) sug­gests a similar process in which "deuterosomes" seem to correspond to the condensation forms. Another possibility is that the preexisting centri­oles may be decondensed into the fibrillar materi­al which would function later as templates for new microtubule proteins (Dirksen and Crocker 1966; Staprans and Dirksen 1974). In cells at a somewhat later stage of ciliogertesis, complete centrioles possessing nine triplets of microtubules accumulate in the luminal apices (Fig. 30). The cells undergoing ciliogenesis usually possess well-developed microvilli. Glycogen granules are often absent from the type II and cil­iated cells. In the developing Syrian hamster tra­chea the fibrillar material with dense granules can first be detected on the 13th gestational day, and

Page 29: Respiratory System

18 Makito Emura

Fig.26 (Upper/eft). Type II (ciliated) cell on the 14th gesta­tional day. Uranyl acetate and lead citrate, x 22770

Fig.27 (Upper right). Higher magnification of Fig. 26. "Growing condensation forms" (GCF) with hollow center; a solitary condensation form (CF) and a ciliary bud-like tubule (arrow) procentriole (PC). Uranyl acetate and lead citrate, x 47320

Fig. 28 (Below). Type II (ciliated) cell on the 14th gestation· al day. Various structures related to ciliogenesis: fibrogran· ular material (FM); centriole (C); condensation forms (CF); procentrioles (PC). Uranyl acetate and lead citrate, x 47320

Page 30: Respiratory System

Development of Syrian Golden Hamster Tracheal Epithelium 19

Fig.29. Type II cell. Centrioles near maturation and con­densation forms (CF). Uranyl acetate and lead citrate, x 31590

Fig.31. Cilia growing from centrioles in type II cells on the 14th gestational day. Uranyl acetate and lead citrate x 16640

only on the following day do ciliated cells occur. Therefore, it can be supposed that a period of 1 day is sufficient for completion of the successive stages preceding ciliogenesis. From the 14th gestational day onward, ciliogene­sis occurs in an increasing number of the type II cells. On the 1st postnatal day, typical mature cil-

Fig.30. Type II cell. Almost mature centrioles. Note the microtubule triplets in one cross section (arrow) and the centriole-associated vesicles (CV). Uranyl acetate and lead citrate, x 31590

iated cells are frequently observed (Fig. 32). How­ever, ciliogenesis on the 14th and subsequent ges­tational days is not always restricted to type II cells, but also occurs in cells apparently of type I at various stages of differentiation of secretory systems, although much less frequently.

Page 31: Respiratory System

20 Makito Emura

Fig.33 (Left). Dorsal epithelium on the 15th gestational day. Note cell with pale cytoplasm (arrow). Hand E, x 170

Differentiation of Mucous Cells

Light Microscopy. On the 14th gestational day type I and type II cells are prominent in the epi­thelium (Figs.21 and 22). Their cytoplasm often appears vacuolated. The epithelium of the 15th and 16th gestational days resembles that of the 14th gestational day. However, the luminal apices of the type I cells are no longer hemispherical but slightly protruded and round. Although occurring at a low frequency, cells with pale cytoplasm be­tween the luminal surface and nucleus appear in the cranial part of the epithelium (Fig. 33). On the last gestational day, the epithelial cells with pale cytoplasm (Fig.33) are more numerous and the first mature mucous cells are observed (Fig. 34).

Fig.32. Mature ciliated cell on the 1st postnatal day. Uranyl acetate and lead citrate, x 11 640

Fig.34 (Right) . Ventral epithelium, 15th gestational day. A few mucous cells with pale cytoplasm. Hand E, x 170

The number of cells that react positively to PAS sharply increases around the 14th gestational day (Emura and Mohr 1975), and continues to in­crease during the next 4 weeks of the postnatal pe­riod. Notably, even the type III cells react posi­tively to PAS, particularly around the nuclei. After diastase pretreatment, however, the positive PAS reaction disappears from around the nuclei of many cells, especially from those of the basal layer (type III). On the 14th gestational day, very few cells react to PAS following diastase, but around the 16th ges­tational or 1st neonatal day the frequency of such cells increases (Emura and Mohr 1975). In these cells only the cytoplasmic portion between the lu­minal surface and nucleus reacts positively to

Page 32: Respiratory System

Development of Syrian Golden Hamster Tracheal Epithelium 21

PAS. With progressive development, these areas acquire more and larger granules, expand, and fi­nally occupy the entire cytoplasm above the nu­cleus. Many differentiating type I cells contain PAS-positive material in the cytoplasm above the nucleus. Such material greatly diminishes in cells toward the caudal part of the epithelium (Fig. 35). Few cells stain with alcian blue until the 14th ges­tational day. From this day onward they remain at an average level of 13% of epithelial cells, which is approximately half the average frequency of PAS-positive cells that resist diastase treatment. Throughout these developmental stages, the ma-

Fig. 35 (Above). Ventral epithelium on the 1st postnatal day. Diastase and PAS, x 106

Fig.36 (Middle). Epithelium on the 1st postnatal day, dor­sal surface. Cells positive to alcian blue are dark. Alcian blue, x 105

Fig.37 (Lower left). Dorsal epithelial cells on the 1st post­natal day. Cytoplasm near lumen stains with alcian blue. Alcian blue and nuclear red, x 425

jority of cells that stain with alcian blue also react to PAS after diastase, but the reverse is not true (Emura and Mohr 1975). On the 14th and 15th gestational days the cyto­plasm of some cells stains with alcian blue around the nucleus and just beneath the luminal surface. These cells occur more frequently in the cranial to middle parts of the dorsal epithelium on the 16th gestational to the 1st postnatal day (Figs.35 and 36). At these stages, cells stain with alcian blue, mainly in the cytoplasmic portion between the lu­minal surface and the nuclei (Figs. 36 and 37). No marked difference can be detected in the fre­quency of cells reacting to PAS and alcian blue

Fig.38 (Lower right). Tracheal epithelium on the 1st post­natal day. On the left is a ciliated cell; in the center are two cells which, following diastase, have PAS-positive material in their cytoplasm. The cell in the center is ciliated; two on the right have spiny processes on the luminal side. PAS and hematoxylin, x 425

Page 33: Respiratory System

22 Makito Emura

Fig.39 (Upper left). Hamster trachea, 13th gestational day. Type I epithelial cell with partly rough vesicles resembling ER. Uranyl acetate and lead citrate, x 11640

Fig.40 (Lower). Differentiating type I cells on the 14th ges­tational day. Cells at two different stages of differentiation. In the cell MCll, the winding cisternae of rough ER which contain dense material are prominent. There already exists a small amount of vesicular ER. The cells MCI seem to be at the same stage as those in Fig.41. The extensions and the local cisternal dilation of the nuclear envelope are no long-

er detectable in the cell Men, while in Mel tlley are both conspicuous (arrows). Note the partly rough membrane portion of the vesicles in Mel. The cell Men apears to be at a later stage of differentiation than the cell Mel. Uranyl acetate and lead citrate, x 11640

Fig.41 (Upper right). Type I (mucous) cells on the 14th ges­tational day. Vesicles are enclosed by a partially granular membrane. Note chromatin condensation, focal cisternal dilation of the nuclear envelope, and glycogen granules (G). Uranyl acetate and lead citrate, x 11640

Page 34: Respiratory System

Development of Syrian Golden Hamster Tracheal Epithelium 23

between the prenatal and the neonatal epithelium on the 16th day. Some ciliated cells on the 16th gestational and the 1st postnatal days clearly react to PAS even after diastase pretreatment (Fig. 38). A few also stain with alcian blue.

Electron Microscopy. In type I cells rough ER no­ticeably increases on the 14th -16th gestational days. It can therefore be presumed that type I cells eventually become mucous cells. However, the ul­trastructure of the rough ER has several different aspects, depending on the stage of differentiation of the cells. In some of the primitive type I cells at the primary stage (Figs.39 and 41), the vacuoles and vesicles possessing both smooth and rough areas begin to resemble ER. These structures are found in blastic nerve cells (Pannese 1968), in de­veloping pancreatic cells (Wessells and Evans 1968), and in embryonic choroidal epithelial cells (Birge and Doolin 1974). The nuclear envelope of these primitive type I cells usually undergoes local cisternal dilation and/ or circumscribed extensions of the outer membrane. This is most frequent and prominent on the 14th gestational day. In differentiating type I cells at advanced stages, almost all of the ER is rough and its flattened winding cisternae are extended and contain electron-dense, fibrillar, and amorphous material (Fig.40). In these type I cells at more advanced stages, rough and smooth vesicular ER occur in addition to the flattened, winding ER. The cisternae of this vesicular ER usually contain fibrillar or amorphous material. Thus, in time, an increasing number of type I cells with cisternae of rough ER and/or vesicles lim­ited by smooth and rough ER membranes, all containing electron-dense, fibrillar, and amor­phous material, fill the entire cytoplasm (Fig. 40). At the same time, dictyosomes of Golgi apparatus develop extensively (Figs.40 and 42). Finally, on the 1st postnatal day, maturing mucous (type I) cells appear (Figs.43 and 44). In these cells the nuclear chromatin is more condensed than that of the mature ciliated cell (Fig. 32). The cisternae of the nuclear envelope are usually locally dilated in many areas (Fig. 44), and the outer nuclear membrane frequently comes into contact with the cytoplasmic vesicles. Some vesi­cles contain less electron-dense material than oth­ers. Both types of of vesicles frequently fuse to­gether in the luminal cytoplasm (Fig. 44). This kind of mucous cell is presumably of the neonatal type, since it occurs only during the neonatal peri­od and is not found in the adult epithelium. On the 1st postnatal day, cells with well-developed

rough ER and Golgi apparatus at various stages of differentiation are still abundant (Fig. 45). From the 14th gestational up until the 1st postna­tal day, many of the epithelial cells, including bas­al cells, contain glycogen granules (Figs.40 and 42). Frequent association of glycogen granules with protrusions of the outer nuclear membrane would suggest that some of these membranes play a part in glycogen metabolism similar to the func­tion of smooth ER of liver parenchymal cells (Coimbra and Leblond 1966). After the 15th gestational day, another type of cell which appears to be secretory in nature occurs (Figs.43 and 46), although its origin is unclear. The cells lack the typical rough ER that synthe­sizes secretory protein. Instead, they contain vac­uoles and vesicles surrounded by smooth and rough membranes. As observed by light micros­copy, after the 14th gestational day cells frequent­ly undergo ciliogenesis (type II) and produce mu­cus (type I). In prospective mucous cells, smooth- and rough­membrane-bound vesicles exist, together with proliferating centrioles (Fig.48). Other such cells possess several cilia, Golgi apparatus, and rough ER, which develop to a considerable extent (Fig.47). In cells which basically resemble the se­cretory cells shown in Fig.46 and which lack granular ER, large smooth- and rough-surfaced vesicles are found together with cilia or proliferat­ed centrioles (Fig.49). However, it is usual in all these types of cells to find that the centrioles, cilia, rough ER, and cytoplasmic vesicles develop poor­ly compared with the same organelles in differen­tiating type I or type II cells. In the mature mu­cous cells no centrioles or cilia can be detected. Cells containing both mucous granules and cilia have been reported in the respiratory epithelium of neonatal rats (Stockinger and Cireli 1965), adult Syrian hamsters in regeneration (McDowell et al. 1979), and adult humans (McDowell et al. 1978).

Changes in Mitotic Activity. In the longitudinally cut hamster tracheal epithelium, the mitotic in­dices are counted in percentages (see Emura and Mohr 1975). The highest percentage of 4.50/0 is seen on the 11 th gestational day during the predif­ferentiation stage. The index sharply declines on the 12th gestational day (2.5%), and then con­tinues to decline until the 14th gestational day (1.8%), when the first ciliated cells can be ob­served. Following this, a more gradual decrease is noticed on the 1st postnatal day (0.3%), when ma­ture mucous cells are first observed.

Page 35: Respiratory System

24 Makito Emura

Page 36: Respiratory System

Development of Syrian Golden Hamster Tracheal Epithelium 25

<1 Fig.42 (Above). Luminal cytoplasm of a differentiating type I cell on the 16th gestational day. Vesiculated ER cis­ternae limited by a rough membrane are especially promi­nent in the cytoplasm near the lumen. Note the well-devel­oped Golgi apparatus and the vesiculated, partly rough ER cisternae. Uranyl acetate and lead citrate, x 22770

Fig.43 (Below). Epithelium, 1st postnatal day, dorsal aspect. Cells in various stages of differentiation. Uranyl acetate and lead citrate, x 5070

Fig. 44 (Above). Type I cell on 1st postnatal day. Nearly ma­ture mucous cell with vesicles of low and high density. In vesicles of high density the limiting membrane is difficult to distinguish. Note the fusion of the two different types of vesicles (arrows). Golgi apparatus (GA). Uranyl acetate and lead citrate, x 16640

Fig.45 (Below). Type I cells in dorsal cranial epithelium on 1st postnatal day. The cell MCIV seems to be at the same stage as that of the cell in Fig.42. The cell MCVI appears to be developing into the cell shown in Fig. 44. Uranyl acetate and lead citrate, x 11640

Page 37: Respiratory System

26 Makito Emura

Fig.46 (Upper left). Secretory cell on the 1st postnatal day. Uranyl acetate and lead citrate, x 11640

Fig.47 (Lower left). A differentiating type I cell (left) and a cell between a mucous and ciliated cell (right), on the 1st postnatal day. Note that the state of the chromatin conden­sation of the cell on the right appears to be at a stage be­tween the ciliated and the type I cell. Uranyl acetate and lead citrate, x 11 640

Uneven Differentiation Pattern of Ciliated and Mu­cous Cells. The first ciliated cells occur in the dor­sal epithelium (pars membranacea) of the trachea on the 14th gestational day. Ciliated cells are vir­tually absent in the ventral and lateral epithelium

Fig.48 (Upper right). A cell in the stage between ciliogene­sis and mucogenesis on the 14th gestational day. Centrioles and partly rough vesicles are prominent. Uranyl acetate and lead citrate, x 7740

Fig.49 (Lower right). A cell in a stage of differentiation be­tween the ciliated cell and the presumed secretory cell in Fig.46. The chromatin condensation is similar to that of the ciliated cell (Fig.32). Uranyl acetate and lead citrate, x 11640

(pars cartilaginea), and at this stage occur more frequently and are longer in the cranial than in the caudal part of the dorsal epithelium. On the 15th gestational day, the cells also appear in the ventral and lateral epithelium and thereafter increase

Page 38: Respiratory System

Epithelial Alterations in Explant Cultures of Fetal Tracheae of Syrian Golden Hamsters 27

gradually in number. Even on the 1st postnatal day they are seen more frequently in the dorsal than in the ventral and lateral epithelium. The cells which are resistant to diastase treatment and react positively to PAS and those cells that stain with alcian blue are unevenly distributed along the ventral and dorsal epithelium during the developmental stages (Figs.35 and 36). Both of these cell types occur primarily in the cranial part of the epithelium. As development progresses, they gradually appear in the caudal part of the ep­ithelium. From the 14th to the 16th gestational days, they occur more frequently in the dorsal than in the ventral epithelium. On the 1st post­natal day, this difference is no longer evident.

References

Anderson RGW, Brenner RM (1971) The formation of basal bodies (centrioles) in the rhesus monkey oviduct. J Cell BioI 50: 10-34

Birge WJ, Doolin PF (1974) The ultrastructural differentia­tion of the endoplasmic reticulum in choroidal epithelial cells of the chick embryo. Tissue Cell 6: 335-360

Coimbra A, Leblond CP (1966) Sites of glycogen synthesis in rat liver cells as shown by electron microscope radio­autography after administration of glucose-H3. J Cell BioI 30: 151-175

Dirksen ER, Crocker IT (1966) Centriole replication in differentiating ciliated cells of mammalian respiratory epithelium. An electron microscopic study. J Microscop 5: 629-644

Emura M (1978) Morphological studies on the develop­ment of tracheal epithelium in the Syrian golden ham­ster. IV. Electron microscopy: blebbing of nuclear mem­brane. Z Versuchstierkd 20: 163-170

Emura M, Mohr U (1975) Morphological studies on the development of tracheal epithelium in the Syrian golden hamster. I. Light microscopy. Z Versuchstierkd 17: 14-26

Frisch D, Farbman AI (1968) Development of order dur­ing ciliogenesis. Anat Rec 162: 221-232

McDowell EM, Barrett LA, Glavin F, Harris CC, Trump BF (1978) The respiratory epithelium. I. Human bron­chus. JNCI 61: 539-549

McDowell EM, Becci PJ, Schurch W, Trump BF (1979) The respiratory epithelium. VII. Epidermoid metaplasia of hamster tracheal epithelium during regeneration fol­lowing mechanical injury. JNCI 62: 995-1008

Mohr U (1973) Effects of diethylnitrosamine on fetal and suckling Syrian golden hamsters. IARC Sci Publ 4: 65-70

Mohr U, Reznik-Schuller H, Emura M (1979) Tissue dif­ferentiation as a prerequisite for transplacental carcino­genesis in the hamster respiratory system, with specific respect to the trachea. Nat! Cancer Inst Monogr 51: 117-122

Pannese E (1968) Developmental changes of the endoplas­mic reticulum and ribosomes in nerve cells of the spinal ganglia of the domestic fowl. J Comp Neurol 132: 331-364

Sorokin SP (1968) Reconstructions of centriole formation and ciliogenesis in mammalian lungs. J Cell Sci 3: 207-230

Staprans I, Dirksen ER (1974) Microtubule protein during ciliogenesis in the mouse oviduct. J Cell BioI 62: 164-174

Stockinger L, Cireli E (1965) Eine bisher unbekannte Art der Zentriolenvermehrung. Z Zellforsch 68: 733-740

Wessells N~ Evans J (1968) Ultrastructural studies of ear­ly morphogenesis and cytodifferentiation in the embry­onic mammalian pancreas. Dev BioI 17 : 413-446

Wi schnitzer S (1974) Die Kernhulle: ihre Ultrastruktur und funktionelle Bedeutung. Endeavour 33: 137-142

Wynder EL, Hecht S (eds) (1976) Lung cancer. In: DICC Technical Report Series, vol 25. International Union Against Cancer, Geneva, pp95-101

Epithelial Alterations in Explant Cultures of Fetal Tracheae of Syrian Golden Hamsters

Makito Emura

Introduction

Various types of epithelial alterations have been observed in explant cultures of differentiated re­spiratory tissues which were treated in vitro with carcinogens (Lasnitzki 1956; Palekar et al. 1968; Crocker and Sanders 1970), infectious micro-

organisms (Gab ridge 1979), mineral dusts (Moss­man et al. 1980), and other airborne particulates (Mossman and Craighead 1979). Explants taken from fetal Syrian golden hamster tracheae (13th-15th day of gestation), which were treated transplacentally with diethylnitrosamine (DEN) (see Table 1) and then cultivated for

Page 39: Respiratory System

28 Makito Emura

Table 1. List of abbreviations

l-APPN l-Acetoxypropylnitrosamine BaA Benz (aJ anthracene BAP N-Nitrosobis (2-acetoxypropyl)amine BeP Benzo(e)pyrene BHP N- Nitrosobis(2-hydroxypropyl)amine BaP Benzo(a)pyrene CHR Chrysene DBN N-Nitrosodibuthylamine DEN N- Nitrosodiethylamine; diethylnitrosamine D HPN 2,2'-Dihydroxy-di-n-propylnitrosamine DMBA 9,10-Dimethyl-l,2-benzanthracene DMDPN N- Nitrosobis(2-methylpropyl)amine DMSO Dimethyl sulfoxide DPN N-Nitrosodi-n-propylamine 2-HPPN N -Nitroso-2-hydroxypropyl-n-propylamine HEPES 4-(2-Hydroxyethyl)-l-piperazineethane sulfonic

M-2-0B MNU MOP MPN N-6-MI NM NMU 2-0PPN PAHs VEN

acid N- Nitrosomethyl(2-oxobutyl)amine Methylnitrosourea N -Nitrosomethyl(2-oxopropyl)amine N- Nitrosomethyl-n-propylamine N- Nitrosohexamethyleneimine N-Nitrosomorpholine Nitrosomethylurea N -Nitroso-2-oxopropyl-n -propyl amine Polycyclic aromatic hydrocarbons N -Nitrosovinylethylamine

4 weeks, showed changes of the same nature as those already observed in vivo. This was also true of explants treated in vitro with polycyclic aro­matic hydrocarbons (PAHs). Epithelium ofthe fe­tal trachea is not fully differentiated at the treat­ment and explant stages: the cells differentiate after several weeks of cultivation. Lesions which are initiated by carcinogens at the beginning of cultivation or at the time of transplacental treat­ment persist within the progeny of affected cells and ultimately are manifest as morphologically discernible alterations.

Tubular Explant Culture of Fetal Tracheae

Two different methods were employed for the treatment and cultivation of fetal tracheal ex­plants. The techniques have already been de­scribed (Emura et al. 1978, 1979; Richter-Reich­helm et al. 1982) but are summarized as follows. Randomly bred, 12-week-old Syrian golden ham­ster~ from the Central Proefdierenbedrijf, Zeist, The Netherlands, were maintained under stan­dard laboratory conditions. The day of mating was regarded as day zero of gestation.

Using the first method, the pregnant females, caged individually, were anesthetized with ether and injected intraperitoneally with 200, 300, or 400 mg DEN per kilogram body wt. on day 13, 14, or 15 of gestation. DEN was dissolved in 1 ml Hanks' solution for injection. Controls received the solvent only. Fetuses were removed by cesar­ean section without direct contact with the moth­er's blood 3Yz-4h after DEN injection. The fe­tuses of treated and control mothers were taken out of the amniotic membranes and rinsed twice in fresh Hanks' solution. The fetal tracheae were dissected under a stereo microscope, divided into cranial and caudal portions, wrapped in chick vi­tellin membrane and cultivated on a membrane filter kept at the gas-medium interface under sta­tionary conditions. Using the second method, tracheal explants of the same gestational time were treated in vitro with the following polycyclic aromatic compounds (PAHs): benzo[a]pyrene (BaP), benz[a]anthracene (BaA), benzo[e]pyrene (BeP), and chrysene (CHR) in quantities of 0.5, 1.5, and 5 mg/ml. Treatment began at 24 h after explantation and continued for 4 days. The cultivation was carried out on a rocker platform moving at 5 cycles per minute. The medium used in both methods was Eagle's minimum essential medium supplement­ed with fetal bovine serum to 20% during the first 4-5 days of cultivation and reduced to 5% there­after. The total cultivation time in both experi­ments ranged from 3 to 9 weeks. The medium was additionally buffered with 20 mM 4-(2-hydroxy­ethyl)-l-piperazineethane sulfonic acid (HEPES).

Alterations After Transplacental DEN Treatment (Method 1)

Hyperplastic proliferation of basal cells (Fig. 50) and squamous metaplasia with (Fig. 51) or with­out cornification were frequently observed. Dys­plasia and metaplasia (Fig. 52) also occurred, par­ticularly in the highest dose group (400 mg/kg body wt.). These alterations can be seen focally in the epithelium, often in more than one site with more than one type of alteration within the same explant. Approximately two-thirds of the total treated explants showed at least one of these alter­ations. Furthermore, metaplastic alterations in­cluding dysplasia occur more frequently than hy­perplasia (a ratio of 5: 1). No mucous cell hyperplasia can be clearly identified. Control ex­plants treated transplacentally with a physiologi-

Page 40: Respiratory System

Epithelial Alterations in Explant Cultures of Fetal Tracheae of Syrian Golden Hamsters 29

cally balanced solution (vehicle for DEN) pro­duce a very well differentiated epithelium with noticeably more ciliated cells than mucous cells (Fig. 53). This is not true, however, of flattened ep­ithelial portions opposite the membrane filter.

Fig. 50 (Above). Hyperplastic growth of cells in the ciliated epithelium of a fetal tracheal explant. Taken after 35 days of cultivation, following transplacental exposure to DEN (200 mg/kg body wt.) on the 13th day of gestation. Note stratified epithelium. Hand E, x 350

Fig.51 (Middle). Squamous metaplasia with cornification in the epithelium of a fetal tracheal explant. Taken after 42 days of cultivation, following transplacental exposure to DEN (200 mg/kg body wt.) on the 14th day of gestation.

Such flattening of the epithelium seems to occur as a result of a shortage of nutrient, the supply of which is only possible through capillary forces in the stationary type of organ culture.

The arrows indicate a vitelline membrane used for this par­ticular organ culture technique. Hand E, x 350

Fig. 52 (Below). Dysplastic epithelium in a fetal tracheal ex­plant after 25 days of cultivation following transplacental exposure to DEN (400 mg/kg body wt) on the 15th day of gestation. The epithelial cells show pleomorphism, cyto­plasmic vacuolization, and disorientation. The arrow indi­cates a vitelline membrane. Hand E, x 350

Page 41: Respiratory System

30 Makito Emura

Alterations After In Vitro Treatment with P AHs (Me~hod2)

Goblet cell hyperplasia (Fig. 54) and epidermoid metaplasia (without cornification) (Fig. 55) are the two alterations most frequently induced by BaP or BaA (up to 5l!g/ml). These alterations are seen focally in the epithelium of approximately one-

Fig. 53 (Above). Epithelium in a control explant of fetal tra­chea excised on the 14th day of gestation and cultivated for 63 days. Ciliated and basal cells can be seen. The lumen is filled with secretion. Hand E, x 350

Fig. 54 (Middle). Mucous cell hyperplasia in the epithelium of a fetal trachea explanted on the 15th day of gestation. Treated in vitro with 5llg/ml BaP and cultivated for

third of the total explants treated with either BaP or BaA, and often occur in more than one site within the same explant. Goblet cell hyperplasia occurs more frequently than epidermoid metapla­sia (a ratio of 3: 1). In the control explants treated with 0.5% dimethyl sulfoxide (DMSO) (solvent for PAHs) and the explants treated with eRR or BaP (up to 5 l!g/ ml), the frequency of squamous

28 days. Note focal proliferation of basal cells (arrows). H and E, x 350

Fig.55 (Below). Squamous metaplasia without cornifica­tion in the epithelium of a fetal trachea explanted on the 15th day of gestation. Treated in vitro with 5llg/ ml BaP and cultivated for 28 days. Hand E, x 350

Page 42: Respiratory System

Epithelial Alterations in Explant Cultures of Fetal Tracheae of Syrian Golden Hamsters 31

metaplasia is low. Mucous cell hyperplasia can­not be clearly identified.

Conclusion

The proliferative and metaplastic alterations seen in explant cultures of fetal Syrian hamster tra­cheae resemble closely those so far observed in other organ culture systems (Marchok et al. 1975), animal models (Mohr et al. 1966; Gould et al. 1971; Harris et al. 1972; Becci et al. 1978), and hu­mans (Trump et al. 1978). A similarity between the focal proliferation of basal cells seen in the BaP­induced goblet cell hyperplasia and that observed in the regenerating hamster epithelium can be seen with light microscopy (McDowell et al. 1979). Recent evidence indicates that mucous cells playa dominant role in the cell proliferation of regenerating hamster tracheal epithelium (Keenan et al. 1982). In explants treated in vitro with BaP or BaA the epithelium underwent no hy­perplasia of ciliated cells or squamous metaplasia with cornification. Both of these alterations occur in explants treated transplacentally with DEN. This can be explained partly by the fact that the first method of explant cultivation promotes dif­ferentiation of ciliated cells rather than mucous cells and the second method does the reverse (Emura et al. 1978, 1979). An auto radiographic study of untreated control explants cultivated by the second method has shown that epithelial cells (indifferent cells, basal cells, and some mucus-containing cells) are la­beled with 3H-thymidine (5 Meilml for 20 min of exposure) at a rate of 19% after 12 h, 14% after 24 h, and 2% after 7 days of cultivation. Although this decreasing tendency of3H-thymidine labeling during cultivation time may partly reflect the in­herent mitotic activity of fetal epithelial cells (Emura and Mohr 1975), the high mitotic rates at the beginning of cultivation can be attributed to the effects of explantation and in vitro cultiva­tion. In such mitosis-accelerating in vitro conditions the susceptibility of cells to carcinogens may well be very different from that encountered in vivo. It seems, therefore, that the difference in treatment and cultivation methods would lead to a variety of induced alterations, although the different na­tures of the carcinogens must also be consid­ered. Transplacental induction of tracheal epithelial tu­mors with DEN is a unique system for the study of respiratory carcinogenesis, in that this particu-

lar organotypic carcinogen (DEN) can affect fe­tal, not fully differentiated, tracheal epithelial cells (Mohr et al. 1966). Tumor development is usually preceded by the occurrence of hyperplas­tic and metaplastic alterations, and there appears to exist a certain correlation between the in vivo and in vitro processes in the occurrence of such early alterations. Therefore, this in vitro system can be used for the identification of fetal cell types susceptible to DEN.

References

Becci PJ, McDowell EM, Trump BF (1978) The respira­tory epithelium. IV. Histogenesis of epidermoid meta­plasia and carcinoma in situ in the hamster. JNCI 61: 577-586

Crocker IT, Sanders LL (1970) Influence of vitamin A and 3,7-dimethyl-2, 6-octadienal (citral) on the effect of ben­zo(a)pyrene on hamster trachea in organ culture. Cancer Res 30: 1312-1318

Emura M, Mohr U (1975) Morphological studies on the development of tracheal epithelium in the Syrian golden hamster. I. Light microscopy. Z Versuchstierkd 17: 14-26

Emura M, Richter-Reichhelm HB, Mohr U (1978) Epithe­lial alterations in fetal tracheal explants of Syrian golden hamsters exposed to diethylnitrosamine in utero. Can­cer Lett 5: 115-121

Emura M, Richter-Reichhelm HB, Emura KM, Matthei S, Mohr U (1979) Tubular explant culture of fetal Syrian golden hamster tracheae. Exp Pathol17: 196-199

Gabridge MG (1979) Hamster tracheal organ cultures as models for infection and toxicology studies. Prog Exp Tumor Res 24: 85-95

Gould VE, Wenk R, Sommers SC (1971) Ultrastructural observations on bronchial epithelial hyperplasia and squamous metaplasia. Cancer 28: 426-436

Harris CC, Sporn MB, Kaufman DG, Smith JM, Jackson FE, Saffiotti U (1972) Histogenesis of squamous meta­plasia in the hamster tracheal epithelium caused by vitamin A deficiency or benzo( a}pyrene-ferric oxide. JNCI 48: 743-761

Keenan KP, Combs JW, McDowell EM (1982) Regenera­tion of hamster tracheal epithelium after mechanical in­jury. III. Large and small lesions : comparative stathmo­kinetic and single pulse and continuous thymidine labelling autoradiographic studies. Virchows Arch (Cell Pathol) 41: 231-252

Lasnitzki I (1956) The effect of 3-4-benz-pyrene on human foetal lung grown in vitro. Br J Cancer 10: 510-516

Marchok AC, Cone MV, Nettesheim P (1975) Induction of squamous metaplasia (vitamin A deficiency) and hyper­secretory activity in tracheal organ cultures. Lab Invest 33: 451-460

McDowell EM, Becci PJ, Schiirch W, Trump BF (1979) The respiratory epithelium. VII. Epidermoid metaplasia of hamster tracheal epithelium during regeneration fol­lowing mechanical injury. JNCI 62: 995-1008

Mohr U, Althoff J, Authaler A (1966) Diaplacental effect

Page 43: Respiratory System

32 Makito Emura

of the carcinogen diethylnitrosamine in the golden ham­ster. Cancer Res 26: 2349-2352

Mossman BT, Craighead JE (1979) Use of hamster tra­cheal organ cultures for assessing the cocarcinogenic ef­fects of inorganic particulates on the respiratory epithe­lium. Prog Exp Tumor Res 24: 37-47

Mossman BT, Adler KB, Craighead JE (1980) Cytotoxic and proliferative changes in tracheal organ and cell cul­tures after exposure to mineral dusts. In: Brown RC, Gormley IP, Chamberlain M, Davis R (eds) The in vitro effects of mineral dusts. Academic, London, pp 241-250

Palekar L, Kuschner M, Laskin S (1968) The effect of.

3-methylcholanthrene on rat trachea in organ culture. Cancer Res 28: 2098-2104

Richter-Reichhelm HB, Emura M, Althoff J (1982) Scan­ning electron microscopical investigations on the respi­ratory epithelium of the Syrian golden hamster. IV. In vitro effects of dimethylsulphoxide and benzo(a)pyrene. Zentralbl Bakteriol Mikrobiol Hyg (B) 176: 269-276

Trump BF, McDowell EM, Glavin F, Barrett LA, Becci PJ, Schiirch W, Kaiser HE, Harris CC (1978) The respirato­ry epithelium. III. Histogenesis of epidermoid metapla­sia and carcinoma in situ in the human. JNCI 61: 563-575

Page 44: Respiratory System

NEOPLASMS

Response to Carcinogens of Respiratory Epithelium, Syrian Golden Hamster (Mesocricetus Auratus)

H.-B. Richter-Reichhelm, W. Boning, and 1. Althoff

Synonyms. Papilloma; epithelial papilloma; squamous cell papilloma; epidermoid papillary tumor; polyp; adenomatous polyp; mixed polyp; polypoid tumor; papillary polyp; mucoepider­moid papillary tumor.

Gross Appearance

Early changes that may lead to neoplasia, such as focal metaplasia with loss of ciliated epithelium and epidermoid metaplasia, hyperplasia and/or papillary dysplasia, cannot be observed macro­scopically, whereas progressively exophytic grow­ing papillary tumors, whether or not they fill the airway lumen, are easily detectable under a mag­nifying glass or a stereo microscope (Fig. 56). In addition to their typical form and shape (sessile or

pedunculated), the tumors are soft and reddish gray, clearly distinguishable from the surrounding respiratory epithelium. Often such areas with small tumors are recognized by increased vascu­larization. Mter perfusion of the tissues with fixa­tive solution, these neoplasms become a yellowish gray contrasting with the whitish adjacent epithe­lium.

Microscopic Features

Nonneoplastic Alterations and Precursor Lesions

Since differentiation of the pseudostratified respi­ratory epithelium in the Syrian golden hamster is not complete at birth, lack of ciliated cells at an early stage of the animals life may represent a

Fig.56. Papillary mucoepidermoid tumor, trachea of Syrian golden hamster, filling almost 50% of the lumen. This neo­plasm was easily detectable with a dissecting microscope. SEM, x 240

Page 45: Respiratory System

34 H.-B.Richter-Reichhelm, W.Boning, and lAlthoff

physiological status rather than pathological change. In terms of surface characteristics of the neonatal epithelium, proliferative patterns prevail during the first 10 days. Less differentiated cells (probably precursors of mucous cells) and mucus­producing cells outnumber ciliated cells for the first 20 days after birth. Time of maturation of the epithelium in the trachea may vary between dif­ferent topographical sites such as the pars mem­branacea, intercartilaginea, and cartilaginea. Dif­ferentiation of epithelium in the respiratory tract appears to be complete approximately 30 days postpartum (Richter-Reichhelm et al. 1980). If criteria for early alterations or precursor lesions are to be specific, non-compound-related degen­erative and regenerative changes of the respirato­ry epithelium have to be recognized and consid­ered in interpretations (McDowell et al. 1979; Althoff et al. 1981). Loss of cilia from the surface of the respiratory epithelium (simple metaplasia) develops due to acute or chronic inflammatory stimuli. Treatment with carcinogens may also cause traumatic injury at the site of application, resulting in such reac­tions. Thus metaplasia together with slight atypia and hyperplasia occur with conversion into a multilayered epithelium. These changes may also appear in regeneration and early neoplastic growth, which may be difficult or impossible to distinguish morphologically. Metaplastic and hyperplastic changes in the respi­ratory epithelium may occur focally, with a marked, well-circumscribed border, or multifocal­ly, less defined and/or disseminated. Simple me­taplasia of the ciliated, pseudostratified epitheli­um lining the airways is easily recognized by loss of cilia and flattening of the epithelial cells. In the cuboidal unilayered epithelium, mucous granules and cilia are only occasionally found; cell nuclei are round or oval and mitotic activity is usually in­creased.

Hyperplasia. The lesions may grow into a multi­layered pattern by an increase in the number of basal cells (so-called basal cell hyperplasia). Less differentiated cells constitute the intermediate layer between the basal cells and surface lining. These cells may be either indifferent or highly dif­ferentiated (e.g., mucous cell hyperplasia).

Squamous Metaplasia. Surface cells are flattened and have horizontally orientated nuclei (parallel to the basal lamina); they may be nonkeratinized or contain keratohyaline granules. In these le­sions, changes such as acanthosis, parakeratosis,

hyperkeratosis, and dyskeratosis may be associat­ed with squamous metaplasia. Desquamation of surface cells is often observed.

Dysplasia; Carcinoma In Situ. Circumscribed areas with a slightly elevated, dome-shaped sur­face are composed of cells with irregular shapes and sizes that proliferate toward the lumen (Fig. 57). In profile, they are irregular in shape and folded. The hyperplastic lesion consists of one or two layers of basal cells (term used in light mi­croscopy) covered by four or five layers of less differentiated (intermediate) cells, sometimes with increased mitotic activity and atypism. Loss of orientation and organization of cells is evident. In cross sections of the epithelium, varying cell shapes are seen; folds appear as papillary projec­tions into the lumen. The uppermost luminal layer is made up either of flat squamous or mucus-pro­ducing cells. Single, ciliated cells occur occasion­ally. The pleomorphism of the cells and the irreg­ularity in cell surface patterns are indicative of dysplasia (Fig. 58) (carcinoma in situ) (Becci et al. 1978).

Papillary Tumors

Exophytic papillary neoplastic alterations of the respiratory epithelium have largely the same cell surface pattern (i. e., epidermoid, less differentiat­ed mucus-producing, and ciliated cells) as those seen in dysplasia (Fig. 59). The epithelial portion of the tumors is supported by a delicate vascular­ized mesenchymal stroma (Fig. 60). Secondary in­flammatory changes are frequent. The basallami­na is seen in all branches of the papillary exophytic growth and is generally not disrupt­ed. If the epithelium of such a papillary neoplasm is composed solely of squamous cells, it may be re­garded as an epidermoid papillary tumor. Sec­tions reveal a flattened layer of cells with horizon­tally oriented nuclei (parallel to the basal mem­brane). Keratin may be formed in some instances. A papillary mucoepidermoid tumor consisting of both mucus-producing and epidermoid cells (Fig. 61) is comparatively easily distinguished by applying Kreyberg stain (mucus = green, kera­tin = red) (Kreyberg 1967). In addition to the pres­ence of differentiated mucus-producing cells at the tumor surface and intraepithelial cysts, cells containing mucous granules are found in the in­termediate zone. Mitoses are seen in all cell layers, although they are not very frequent. Subepithelial

Page 46: Respiratory System

Response to Carcinogens of Respiratory Epithelium, Syrian Golden Hamster 35

Fig.57 (Above). Circumscribed area of trachea consisting of cell surfaces varying in shape and form indicating hy­perplasia and dysplasia. Note folds and slight projection into the tracheal lumen. SEM, x 320

mucous glands may also undergo metaplasia and proliferative changes, a feature which could be falsely interpreted as invasive growth. In addition, papillary tumors may originate in the cystic ducts of the subepithelial glands.

Fig.58 (Below). Dysplastic alteration (carcinoma in situ) of the respiratory epithelium, trachea. Note multilayered aty­pical cell arrangement with vacuoles and epidermoid cells in the uppermost layer. Semithin section, toluidine blue, x 350

Ultrastructure

Nonneoplastic and Precursor Lesions

Simple Metaplasia. In simple metaplasia, cells are marked by the loss of cilia and intracytoplasmic ciliary bodies. Fewer cilia than normal may occur in some cells and occasionally cells with cytoplas­mic protrusions may contain cilia. In cross sec-

Page 47: Respiratory System

36 H.-B. Richter-Reichhelm, W. Boning, and 1. Althoff

Fig.59 (Above). Early mucoepidermoid papillary tumor, trachea, with intraepithelial cysts and focal squamous met­aplasia. Note the delicate vascularized connective tissue stalk (S). In some areas under the basal lamina early tumor growth seems to interrupt the basement membrane (ar­rows). Semithin section, toluidine blue, x 150

tions of these cilia, micro filamentous patterns ap­pear to be normal, with nine doublets around the periphery and two in the center. In some in­stances, degenerative changes such as swelling of mitochondria and multivesiculated and laminat­ed bodies (secondary Iysosomes) may be found. The luminal cell surface bears short and numer­ous microvilli; cell borders are clearly distinguish-

Fig.60 (Lower left). Mucoepidermoid papillary tumor of the respiratory epithelium filling the tracheal lumen. Note delicate mesenchymal stroma with capillaries (S). Hand E, x 80

Fig.61 (Lower right). Epithelial cells, trachea, containing mucus (M) (green in Kreyberg stain). At the apex (a), flat­tened cells with reddish cytoplasm indicate nonkeratinized squamous cells. Kreyberg stain, x 440

able by the slightly elevated cell membrane and increased density of cytoplasmic protrusions. Generally, all cells have a similar shape and size and nuclei are oval and uniform.

Squamous Metaplasia. In epidermoid metaplasia of the respiratory epithelium, the alignment of cellular and nuclear axes parallel to the basal

Page 48: Respiratory System

Response to Carcinogens of Respiratory Epithelium, Syrian Golden Hamster 37

membrane is easily recognizable. Many cells in the now multilayered epithelium contain bundles of tonofilaments. Keratin granules are not always detectable. The number of desmosomes has large­ly increased and interdigitations of cell mem­branes are pronounced. In some areas of epider­moid metaplasia, nuclei are flat and deeply invaginated.

Hyperplasia. In a thick epithelium, with general loss of cilia at the luminal cell surface, ciliogenesis may be disturbed as cells appear to produce premature intracytoplasmic cilia. These cilia may be found even at infranuclear sites close to the basal epithelial layers. In other areas, less differ­entiated and highly differentiated mucous cells may compose the majority of cells in focal lesions (mucous cell hyperplasia). These cells have highly developed and conspicuous endoplasmic reticu­lum, as well as Golgi complexes. Large mucous granules may not be present in all cells.

Dysplasia. Ultrastructural features of epithelial cells in sites of cellular atypia include empty vacu­oles, intraepithelial cysts filled with mucus, and occasionally structurally intact cilia. These fea­tures lead to the diagnosis of dysplasia or carcino­ma in situ. Varying degrees of cellular differentia­tion form the epithelial pattern, particularly of the surface structure. On some occasions, dense core granules, as seen in neurosecretory cells or

"amine precursor uptake and decarboxylation" (APUD) cells, are formed. Small areas can be seen which contain squamous cells at the irregu­larly folded luminal surface of the epithelium. Cell nuclei vary in size and form; they often con­tain deep invaginations and atypical mitotic fig­ures.

Papillary Tumors

The structure, seen by surface electron micro­scopy, of papillary tumors arising from the respi­ratory epithelial lining may have varying patterns. If the surface of the tumor cells is covered with patterns arranged in microfolds and microridges, characteristic of nonkeratinized squamous cells, the diagnosis of epidermoid tumor can be made. Frequent mitoses support the proliferative nature of this exophytic neoplastic growth. In some in­stances desquamation of flat surface cells may be prominent, exposing the surface of underlying less differentiated cells. Some papillary tumors have the features of poly­morphism similar to that seen in dysplasia. In ad­dition to the less differentiated areas consisting of cells with varying numbers of microvilli, groups of well-differentiated cells with cilia and mucus are found (Fig. 62). Ciliogenesis and mucogenesis may not be restricted to the surface layer (Figs. 63 and 64). In other parts of the same tumor, numer­ous desmosomes may be found, and some cells

Fig. 62. Early papillary tumor, trachea. The surface of the mass consists of squamous mucus-producing and ciliated cells. SEM, x 420

Page 49: Respiratory System
Page 50: Respiratory System

Response to Carcinogens of Respiratory Epithelium, Syrian Golden Hamster 39

contain increased numbers of bundled filaments. Cell boundaries are digitated, nuclei are flat and invaginated (Fig. 65). Keratohyaline granules may be found occasionally and sometimes cells situat­ed in the intermediate layer contain neurosecreto­ry granules. Intracytoplasmic vacuoles and in­traepithelial cysts are frequent. Usually, the basal lamina is intact throughout the entire tumor stalk. In these papillary tumors, mitotic figures may be greater in number than in dysplasia.

Differential Diagnosis

Papillary tumors of the surface respiratory epithe­lium (nasal cavities, larynx, trachea, bronchi) are classified according to their epithelial component, i. e. neoplasms with squamous epithelium on their surface (nonkeratinizing or keratinizing) are clas­sified as epidermoid (or squamous) papillary tu­mors. Exophytic neoplasms which contain goblet cells, an occasional ciliated cell, and squamous cells should be named mucoepidermoid papillary tumors. These tumors may also be made up of areas of less differentiated cells. Their exophytic growth is usually noninvasive. When infiltration occurs, the neoplasms gain characteristics of epidermoid or mucoepidermoid carcinomas. Routine histology (hematoxylin-eosin or toluidine blue stain) may render sufficient information in terms of neoplas­tic growth, but only special stains such as PAS reaction, alcian blue, and/or Kreyberg's method (Kreyberg 1967) allow the cytological identifica­tion of cell status necessary for differential diag­nosis (Herrold 1964a; Herrold and Dunham 1963; Dontenwill and Mohr 1961, 1962; Mohr 1970). Transmission electron microscopy (TEM) and scanning electron microscopy (SEM) prepa­rations may provide evidence of histiogenesis, es­pecially when the tissue is less differentiated.

<l Fig. 63 (Above). Mucus-producing cell from a mucoepider­moid respiratory tumor, bronchus, hamster. Note the Golgi complex (G) and mucous granules (M). x 47500

Fig.64 (Middle). Mucoepidermoid respiratory tumor, bronchus, revealing multiple ciliary bodies (C) in a cell at­tached to the basal lamina. TEM, x 9500

Fig.65 (Below). Portion of a respiratory mucoepidermoid tumor, bronchus, in a flattened cell layer. Nuclei are digi­tated (N), cytoplasm contains bundled tonofilaments (E). Note abundant desmosomes (D). TEM, x 6900

Biologic Features

Papillary tumors of the larynx, trachea, and bron­chi develop as single or multiple neoplasms. They often originate in the epithelium of the membran­aceous portion if cartilage partially composes the airway. The tumors sometimes occlude the lumen, causing death by suffocation. Neoplasms that partially close the airway lumen can cause irregu­lar breathing. Distal portions of the respiratory tract may undergo atelectases and emphysema, and infections of the lung enhanced by hampered clearance and defense mechanisms often occur. Papillary tumors arising in the respiratory epithe­lium of the larynx, trachea, and bronchi may be tom from their site of origin and implanted into the lung after aspiration (Sellakumar et al. 1976). This could easily be the case if the neoplasms are pedunculated and treatment by intralaryngeal and intratracheal instillation techniques is contin­ued after tumors have already occurred. In general, metastasis of these papillary tumors of the epithelial lining of the respiratory tract do not occur frequently. Only occasionally is invasive growth found in the mediastinum (Herrold 1964b; Althoff et al. 1977). Papillary tumors also arise in the ducts of subepithelial glands. Since the epithelium of these glands also undergoes metaplasia, difficulties may occur in diagnosing true invasive growth. They may be malignant, since they are transplantable into other tissues. The respiratory epithelium of the Syrian golden hamster is highly sensitive to the carcinogenic ef­fects of systemically acting compounds as well as to locally applied agents. The early and advanced stages of papillary tumors described here have de­veloped after exposure to a carcinogen (nitros­amines, polycyclic aromatic hydrocarbons). Rela­tively few tumors of the larynx, trachea, and bronchi have been reported in untreated hamsters (Pour et al. 1976a; Homburger et al. 1983).

Comparison with Other Species

Compared with other laboratory animals, spon­taneous pathological alterations and neoplastic changes of the respiratory epithelium in Syrian golden hamsters are infrequent (Nettesheim 1972; Pour et al. 1976a; Mohr and Richter-Reichhelm 1982). Nonneoplastic alterations, caused mainly by infections, are found in all species. Due to spe­cific anatomic conditions, sites where metaplasia occurs more frequently have been described in

Page 51: Respiratory System

40 H.-B. Richter-Reichhelm, W. Boning, and 1. Althoff

man (Auerbach et al. 1962), pigs, dogs, and rab­bits (Wang et al. 1972). Squamous metaplasia is very common at ductal openings of subepithelial glands and bifurcations of the tracheobronchial tree. This phenomenon has been attributed to the heavy deposition of in­haled particles at these particular sites, resulting in intense and prolonged alterations of the func­tion of the mucociliary clearance system. If the results of experimental respiratory tract car­cinogensis are to be interpreted accurately, the rat as an experimental model should be considered with caution. Chronic endemic bronchitis (see p. 211), which results in severe regenerative and dysplastic alterations progressing to overt bron­chiectasis, is commonly found in many strains (Pour et al. 1976b). Very little is known about spontaneous lesions in the main airways of mice. Certain differences exist between tumors of the upper respiratory system (larynx, trachea, bron­chi) in hamsters and humans. In man papillary tu­mors of the larynx are more frequent than those of the trachea, but in hamsters tracheal neoplasms predominate. In the hamster, this reflects the ef­fect of carcinogenesis on a specific anatomic lo­calization. Tumors which mainly occur in young human patients may be multiple or even diffuse, involving the entire tracheobronchial tree. For a long time, single papillary tumors were described as developing predominantly in the stem bronchi and carina, and also occasionally in the main and segmental bronchi (Liebow 1952). From the his­togenetic point of view, the induced epithelial changes described in the respiratory tract of the Syrian golden hamster are quite comparable to findings in human bronchi (McDowell et al. 1982; Melamed and Zaman 1982). Other hamster species, such as the European hamster (Cricetus cricetus L) and the Chinese hamster ( Cricetulus griseus M. E.) have been inves­tigated in studies of respiratory tract carcinogene­sis. The Syrian golden hamster (Mesocricetus au­ratus W) should be regarded as the most appro­priate model, partly because of the knowledge of the physiology and pathology of lesions of this species (Mohr and Reznik 1982).

References Althoff J, Grandjean C, Russell L, Pour P (1977) Vinyl­

ethylnitrosamine: potent respiratory carcinogen in Syrian hamsters. JNCI 58: 439-442

Althoff J, Richter-Reichhelm HB, Green U, Kracke D (1981) Scanning electron microscopical investigations on the respiratory epithelium of the Syrian golden ham­ster. III Regeneration after traumatic injury. Zentralbl Bakteriol (B) 174: 249-259

Auerbach 0, Stout AP, Hammond EC, Garfinkel L(1962) Bronchial epithelium in former smokers. N Engl J Med 267:119-125

Becci PJ, McDowell EM, Trump BF (1978) The respiratory epithelium. IV. Histogenesis of epidermoid metaplasia and carcinoma in situ in the hamster. JNCI 61: 577-586

Dontenwill W, Mohr U (1961) Carcinome des Respira­tionstractus nach Behandlung von Goldhamstern mit Diathylnitrosamine. Z Krebsforsch 64: 305-312

Dontenwill W, Mohr U (1962) Vergleichende Untersu­chungen an metaplastischen und malignen Epithel­veranderungen des Respirationstraktes im Tierexperi­ment. Z Krebsforsch 65: 168-170

Herrold KM (1964a) Epithelial papillomas of the nasal cavity. Arch Pathol78: 189-195

Herrold KM (1964b) Effect of route of administration on the carcinogenic action of diethylnitrosamine (N-nitros­odiethylamine). Br J Cancer 18: 763-767

Herrold KM, Dunham U (1963) Induction of tumors in the Syrian hamster with diethylnitrosamine (N-nitroso­diethylamine). Cancer Res 23 :773-777

Homburger F, van Dongen CG, Adams R, Soto E (1983) Standardizing Syrian hamsters for toxicology. In: Hom­burger F (ed) Safety evaluation and regulation of chemi­cals. Karger, Basel, pp 225-232

Kreyberg L (1967) International histological classification of tumours. No 1: Histological typing of lung tumours. WHO, Geneva

Liebow AA (1952) Tumors of the lower respiratory tract. In: Atlas of tumor pathology, 1st series, Fasicle 17. Armed Forces Institute of Pathology, sect 5, Washington DC

McDowell EM, Becci PJ, Schurch W, Trump BF (1979) The respiratory epithelium. VII. Epidermoid metaplasia of hamster tracheal epithelium during regeneration fol­lowing mechanical injury. JNCI 62: 995-1008

McDowell EM, Harris CC, Trump BF (1982) Histogenesis and morphogenesis of bronchial neoplasms. In: Shi­mosato Y, Melamed MR, Nettesheim P (eds) Morpho­genesis oflung cancer, vol 1. CRC, Boca Raton, pp 1-36

Melamed MR, Zaman MB (1982) Pathogenesis of epider­moid carcinoma of the lung. In: Shimosato Y, Melamed MR, Nettesheim P (eds) Morphogenesis of lung cancer, vol 1. CRC, Boca Raton, pp 37 -64

Mohr U (1970) Effects of diethylnitrosamine in the respi­ratory system of Syrian golden hamsters. In: Nettesheim P, Hanna MG, Deatherage JW (eds) Morphology of experimental respiratory carcinogensis, AEC Sympo­sium Series No 21. USAEC, Division of Technical In­formation Extension, Oak Ridge, pp 255-265

Mohr U, Reznik G (1982) Three hamster species as models in cancer research. In: Turusov VS (ed) Pathology oftu­mours in laboratory animals, vol III. Tumours of the hamster. IARC Sci Pub134: 437-442

Page 52: Respiratory System

Mohr U, Richter-Reichhelm HB (1982) The impact of spontaneous pathology on the quality of animals for toxicological studies. In: Bartosek I, Guaitani A, Pacei E (eds) Animals in toxicological research. Raven, New York, pp 65-70

Nettesheim P (1972) Respiratory carcinogenesis studies with the Syrian golden hamster: a review. Prog Exp Tu­mor Res 16: 185-200

Pour P, Mohr U, Cardesa A, Althoff J, Kmoch N (1976 a) Spontaneous tumors and common diseases in two co­lonies of Syrian hamsters. II. Respiratory tract and di­gestive system. JNCI 56: 937-948

Pour P, Stanton MF, Kuschner M, Laskin S, Shabad LM (1976 b) Tumours of the respiratory tract. In: Turusov VS

Polypoid Adenoma, Nasal Mucosa, Rat 41

(ed) Pathology of tumours in laboratory animals, vol 1. Tumours of the rat, part 2. IARC Sci Pub16: pp 1-61

Richter-Reichhelm HB, Emura M, Althoff J (1980) Scan­ning electron microscopical investigations on the respi­ratory epithelium of the Syrian golden hamster. I. Post­natal differentiation. Zentralbl Bakteriol (B) 171: 424-432

Sellakumar A, Stenback F, Rowland J (1976) Effects of dif­ferent dusts on respiratory carcinogenesis in hamsters induced by benzo( a )pyrene and diethylnitrosamine. Eur J Cancer 12: 313-319

Wang N-S, Huang S-N, Thurlbeck WM (1972) Squamous metaplasia of the opening of bronchial glands. Am J Pa­thol67: 571-582

Polypoid Adenoma, Nasal Mucosa, Rat

William D. Kerns

Synonyms. Adenomatous polyp; papillary adeno­ma; adenoma; papilloma.

Gross Appearance

Polypoid adenomas are usually observed in the most anterior part of the nasal cavity (Kerns et al. 1983a, b). They may vary in size from small mi­croscopic nodules to masses large enough to pro­trude from the nares and may cause dyspnea. While the adenomas are noninvasive, larger ones may cause obstruction of the nasolacrimal duct or

Fig.66. Coronal section of nasal cavity from a rat following exposure to formaldehyde. A small polypoid adenoma (arrows) has arisen on the lateral side of the maxilloturbinate. ND, nasolacrimal duct; N, nasoturbinate; M, maxilloturbinate; V, vomeronasal organ

induce pressure atrophy of adjacent structures. In formalin-fixed decalcified coronal sections, the tumor appears as a solid, whitish gray mass (Fig. 66).

Microscopic Features

Polypoid adenomas may be sessile (Fig. 67) or pe­dunculated (Fig.68). They arise from the mucosa of the nasoturbinates, maxilloturbinates, or lateral wall of the anterior nasal cavity. The lamina pro­pria contains few inflammatory cells and little

Page 53: Respiratory System

42 William D. Kerns

supporting connective tissue, but is well vascular­ized. Nonciliated tumor cells of the adenomas vary morphologically from cuboidal to low co­lumnar epithelium; they form solid sheets or mi­crocysts. In hematoxylin and eosin preparations, the cells are recognized by basophilic cytoplasm and centrally located nuclei. The cysts contain PAS-positive material, sloughed epithelium, and inflammatory cells (Fig. 67). Adenomas appear to originate as hyperplastic, ex­ophytic, mucosal nodules. Origin from the sub­mucosa seems unlikely, since tumors frequently arise in areas that are devoid of submucosal glands and the tumor cells have light and electron microscopic features of respiratory epithelium (Monteiro-Riviere and Popp 1984). Morphologi­cal characteristics of larger tumors suggest that, with continued growth, the surface epithelium in­vaginates into the tumor mass, thus forming mi­crocysts (Fig. 69).

Ultrastructure

Adenomas consist of nonciliated electron-dense (dark) and electron-lucent (light) cuboidal and low columnar epithelial cells (Fig.70). Dark cells can be recognized by their short, nonbranching

microvilli, centrally located convoluted nuclei, small apical vacuoles, basal clusters of mitochon­dria, and electron-dense cytoplasm (Fig. 71). Light cells are also characterized by centrally lo­cated nuclei, but with fewer indentations of the nuclear membrane. Light cell cytoplasm is elec­tron lucent and contains small apical vacuoles and randomly distributed mitochondria. Most but not all light cells have short, nonbranching micro­villi (Fig. 72). The cell membranes of both types of cells interdigitate extensively, and most cells have a single desmosome in their apical junctional zone. Light and dark cells appear to have equal complements of rough endoplasmic reticulum and polyribosomes. Ultrastructurally, the cells comprising polypoid adenomas have the characteristics of respiratory epithelium (Monteiro-Riviere and Popp 1984).

Differential Diagnosis

Polypoid adenomas should pose few diagnostic problems. In some cases, it may be difficult to de­termine whether the cell of origin is of respiratory epithelium or submucosal glandular epithelium. Klaassen et al. (1982) have reported that the sub­mucosal glandular epithelium is characterized

Fig. 67. A sessile polypoid adenoma from the nose of a rat folowing exposure to formaldehyde. Microcysts contain PAS­positive material. Hand E, x 230 (reduced by 15%)

Page 54: Respiratory System

Fig.68 (Above). A pedunculated polypoid adenoma of the mucosa, maxilloturbinate of a rat after exposure to for­maldehyde. Hand E, x 180

Polypoid Adenoma, Nasal Mucosa, Rat 43

Fig.69 (Below). A polypoid adenoma of the nasoturbinate of a rat after exposure to formaldehyde . Apparent invagi­nation (arrows) of the surface mucosa. Hand E. x 360

Page 55: Respiratory System

44 William D. Kerns

Fig. 70. Light (L) and dark (D) cells in a polypoid nasal adenoma in a rat after exposure to formaldehyde. A small cyst filled with neutrophils and debris is present. TEM, x 2500 (reduced by 15%)

Fig.71. A nonciliated dark cell from an adenoma, recognized by its apical vacuoles, basal clusters of mitochondria, and electron-dense cytoplasm. Note elaborate interdigitation of the cell membrane (M), characteristic of nasal respiratory epithelium. TEM, x 8000 (reduced by 15%)

Page 56: Respiratory System

Fig. 72. A non ciliated light cell from an adenoma, recognized by its small apical vacuoles, randomly distributed mitochondria, and electron-lucent cytoplasm. Both light and dark cells usually have short, nonbranching microvilli (M). TEM, x 8000 (reduced by 15%)

ultrastructurally by apical secretory granules (0.5-1.0 ~m) of varying electron density and well­developed rough endoplasmic reticulum. Ultra­structural examination and/ or immunohisto­chemical reactions for keratin (aKl antigen) may be helpful in identifying periacinar myoepithelial cells. Myoepithelial cells and large secretory gran­ules would normally be found only in adenomas originating from the submucosal glands (Nath­rath et al. 1982; Klaassen et al. 1982).

Biologic Features

A polypoid adenoma has been reported, as a spontaneous lesion, in a male Fischer 344 rat (Kerns et al. 1983 b), and this tumor has been ob­served in control and treated rats from toxicity studies of a variety of chemical compounds (Table 2; Stinson 1983). No evidence is available to suggest that polypoid adenomas progress to adenocarcinomas of the nasal cavity in the rat. It has been reported (Taka­no et al. 1982) that focal nodular hyperplasia (in­verted papilloma) is much more important in the

Polypoid Adenoma, Nasal Mucosa, Rat 45

pathogenesis of nasal adenocarcinoma than are polypoid adenomas (exophytic papilloma).

Comparison with Other Species

In humans, epithelial papillomas (also termed na­sal papillomatosis, squamous papilloma, squa­mous papillary epithelioma, Schneiderian papil­loma, cylindric papilloma, or transitional cell papilloma), not adenomas, are the most common benign neoplasms of the nasal cavity. Even so, they are relatively rare lesions, being only 1125th as frequent as the more common inflammatory nasal polyp (Hyams 1971; Lasser et al. 1976; Ri­dolfi et al. 1977; Snyder and Perzin 1972). Mor­phologically similar neoplasms, identified as mi­crocystic papillary adenomas, have been reported in humans, and they represent 1.6% of all tumors of the nose and sinuses (Friedmann and Osborn 1982). Malignant transformation has not been en­countered. Nasal adenomas have been reported in dogs and cats (Madewell et al. 1976), but morphological de­scriptions were not provided. Transmissible ovine

Page 57: Respiratory System

46 William D. Kerns

Table 2. Polypoid adenomas observed in the nasal cavity of the rat

Chemical Strain Male"

Formaldehyde F344 0.0 ppm 1/118 2.0 ppm 4/118 5.6 ppm 6/119

14.3 ppm 4/117

Phenacetin S-D 1.25% 3/22 2.5% 0127

p-Cresidine F344 0.5% 1/48 1.0% 2/45

1.4-Dinitrosopiperazine F344 0.01% 81/125

Hexamethylphosphoramide S-D 100ppb 11200b

Nitrosaminobutanone F344 11.7 mg/ day for 60 days (SC) 4/12c

2,6-Xylidine S-D 1000 ppm 2/56 3000 ppm 10/56

Ethyl acrylate F344 Oppm 1/59 o ppm 0/62

25 ppm 0/77 75 ppm 0178

225 ppm 1170

SD. Sprague-Dawley; ND. not determined a No. of tumors/no. of nasal cavities evaluated b Sex not reported c No. of tumors represents adenomas and papillomas

adenomas, adenopapillomas, and adenocarcino­mas have been reported (Njoku et al. 1978; Yonemichi et al. 1978), but are morphologically distinct from polypoid adenomas of the rat. Adenomas have also been reported in mice (Rez­nik et al. 1980), gerbils (Cardesa et al. 1976), and hamsters (Feron et al. 1982). Adequate morpho­logical descriptions and photomicrographs were not provided and it is not certain if these tumors are similar to those of the rat.

Female" Study type Reference

Inhalation Kerns et al. 0/114 1983a, b 4/118 0/116 1/115

Feeding Isaka et al. 3125 1979 1127

Feeding Reznik et al. 1/46 1981 6/47

Drinking Takano et al. ND water 1982

Inhalation Lee and Trochimowicz 1982

Injection Hecht et al. 6/12c 1980

0/56 Feeding Kornreich and 6/56 Montgomery

1984

1/60 Inhalation 1. Young personal 0/60 communication 1175 1175 0171

References

Cardesa A, Pour P, Haas H, Althoff J, Mohr U (1976) His­togenesis of tumors from the nasal cavities induced by diethylnitrosamine. Cancer 37: 346-355

Feron VJ, Kruysse A, Woutersen RA (1982) Respiratory tract tumours in hamsters exposed to acetaldehyde va­pour alone or simultaneously to benzo(a)pyrene or di­ethylnitrosamine. Eur J Cancer Clin Oncol18: 13-31

Friedmann I, Osborn DA (1982) Tumours of the mucosal glands. In: Friedmann I, Osborn DA (eds) Pathology of granulomas and neoplasms of the nose and paranasal si­nuses. Churchill Livingstone, New York, 133-161

Hecht SS, Chen C-HB, Ohmori T, Hoffmann D (1980) Comparative carcinogeniticy in F344 rats of the tobac­co-specific nitrosamines, N' -nitrosonornicotine and 4-(N-methyl-N-nitrosamino )-1-(3-pyridyl)-1-butanone. Cancer Res 40: 298-302

Hyams VJ (1971) Papillomas of the nasal cavity and para­nasal sinuses. A clinicopathological study of 315 cases. Ann Otol Rhinol Laryngol80: 192-206

Page 58: Respiratory System

Isaka H, Yoshii H, Otsuji A, Koike M, Nagai Y, Koura M, Sugiyasu K, Kanabayashi T (1979)Tumors of Sprague­Dawley rats induced by long-term feeding of phenace­tin. Gan 70: 29-36

Kerns WD, Donofrio DJ, Pavkov KL (1983 a) The chronic effects of formaldehyde inhalation in rats and mice. A preliminary report. In: Gibson JE (ed) Formaldehyde toxicity. Hemisphere, New York, pp 111-113

Kerns WD, Pavkov KL, Donofrio Dl, Gral1a El, Swen­berg lA (1983 b) Carcinogenicity of formaldehyde in rats and mice after long-term inhalation exposure. Can­cer Res 43: 4382-4392

Klaassen ABM, lap PHK, Kuijpers W (1982) Ultrastruc­tural aspects of the nasal glands in the rat. Anat Anz 151: 455-466

Kornreich M, Montgomery CA (1984) Technical report on the carcinogenesis bioassay of 2,6-xylidine (2,6-dime­thylanaline). Charles River C-D rats (diet study). CAS 87-62-7

Lasser A, Rothfeld PR, Shapiro RS (1976) Epithelial papil­loma and squamous cel1 carcinoma of the nasal cavity and paranasal sinuses: a clinicopathological study. Can­cer 38: 2503-2510

Lee KP, Trochimowicz HI (1982) Induction of nasal tu­mors in rats exposed to hexamethylphosphoramide by inhalation. INCI 68: 157-171

Madewel1 BR, Priester WA, Gillette EL, Snyder SP (1976) Neoplasms of the nasal passages and paranasal sinuses in domesticated animals as reported by 13 veterinary col1eges. Am 1 Vet Res 37: 851-856

Monteiro-Riviere N, Popp lA (1984) Ultrastructural char­acterization of the nasal respiratory epithelium in the rat. Am 1 Anat 169: 31-43

Nathrath WB, Wilson PD, Trejdosiewicz LK (1982) Im­munohistochemical localisation of keratin and luminal

Neoplasms, Mucosa, Ethmoid Turbinates, Rat 47

epithelial antigen in myoepithelial and luminal epithe­lial cells of human mammary and salivary gland tu­mours. Pathol Res Pract 175: 279-288

Njoku CO, Shannon D, Chineme CN, Bida SA (1978) Ovine nasal adenopapilloma: incidence and clinico­pathologic studies. Am 1 Vet Res 39: 1850-1852

Reznik G, Reznik-Schiil1er HM, Hayden DW, Russfield A, Murthy ASK (1981) Morphology of nasal cavity neo­plasms in F344 rats after chronic feeding of p-cresidine, an intermediate of dyes and pigments. Anticancer Res 1: 279-286

Reznik G, Ul1and B, Stinson SF, Ward 1M (1980) Mor­phology and sex-dependent manifestation of nasal tu­mors in B6C3F1 mice after chronic inhalation of 1,2-dibromo-3-chloropropane. 1 Cancer Res Clin Oncol 98: 75-83

Ridolfi RL, Lieberman PH, Erlandson RA, Moore OS (1977) Schneiderian papillomas: a clinicopathologic study of 30 cases. Am 1 Surg Pathol1 : 43-53

Snyder RN, Perzin KH (1972) Papillomatosis of nasal cav­ity and paranasal sinuses (inverted papilloma, squa­mous papilloma). A clinicopathologic study. Cancer 30: 668-690

Stinson SF (1983) Nasal cavity cancer in laboratory animal bioassays of environmental compounds. In: Reznik G, Stinson SF (eds) Nasal tumors in animals and man, vol III. Experimental carcinogenesis. CRC, Boca Raton, chap 7

Takano T, Shirai T, Ogiso T, Tsuda H, Baba S, Ito N (1982) Sequential changes in tumor development induced by 1,4-dinitrosopiperazine in the nasal caity of F344 rats. Cancer Res 42: 4236-4240

Yonemichi H. Ohgi T, Fujimoto Y, Okada K, Onuma M, Mikami T (1978) Intranasal tumor of the ethmoid olfac­tory mucosa in sheep. Am 1 Vet Res 39: 1599-1606

Neoplasms, Mucosa, Ethmoid Turbinates, Rat

Sherman F. Stinson and Hildegard M. Reznik-Schuller

Synonyms. Adenocarcinoma; squamous cell car­cinoma; olfactory neuroblastoma, esthesioneuro­blastoma.

Gross Appearance

Neoplasms of the ethmoid regions of the nasal cavities of rats appear, in advanced cases, as large masses exhibiting both exophytic and endophytic patterns of growth. One or both sides of the poste­rior nasal cavities may be completely filled by the tumor, resulting in displacement, erosion, and de­struction of the septum, turbinals, and other sur-

rounding osseous and cartilaginous structures. Extension through the cribriform plate into the ol­factory lobes of the brain can frequently be visual­ized grossly upon longitudinal or coronal section­ing.

Microscopic Features

Three major types of neoplasms have been report­ed to occur in the ethmoid regions of the nasal cavities of rats: adenocarcinomas, squamous cell carcinomas, and olfactory neuroblastomas. The adenocarcinomas are usually poorly differentiat-

Page 59: Respiratory System

48 Shennan F. Stinson and Hildegard M. Reznik-Schuller

Fig.73. Poorly differentiated adenocarcinoma of the eth­moturbinal regional of a F344 rat fed a diet containing p-cresidine for 2 years. Disorganized masses of cells with round to oval nuclei and little cytoplasm are seen. Hand E, x 130

ed and consist of masses of anaplastic cells having scant cytoplasm and large round to oval hyper­chromatic nuclei demonstrating considerable ple­omorphism (Fig. 73). Bizarre mitotic figures are common (Fig.74). The pattern of differentiation varies in different parts of the neoplasms - some areas contain relatively well-differentiated cells and arrangement, while most areas appear less well differentiated with a lack of cellular organi­zation and orientation (Fig. 75). Some lesions have a higher proportion of well-differentiated areas with glandular architecture. Fine fibrovas­cular septa course through the tumors, dividing them into lobules. Most adenocarcinomas con­tain areas with formation of rosettes, pseudoro­settes or small acinar structures. These rosettes are formed by tumor cells, usually poorly differentiat­ed or anaplastic, that are oriented around a cen­trallumen or blood vessel (Figs. 73 and 74). Areas of squamous differentiation are also found within these neoplasms (Fig. 76). Poorly differentiated adenocarcinomas invade into and through the bones surrounding the posterior nasal cavities

Fig.74. Higher magnification of the poorly differentiated adenocarcinoma in Fig. 73. Considerable pleomorphism and a high mitotic rate are in evidence. Organization of cells into rosette structures is seen. Hand E, x 330

(Fig. 77) and through the cribriform plate into the olfactory lobes of the brain (Fig. 78). Areas of in­vasion have architectural and cytological charac­teristics typical of the primary tumor. Metastases occur frequently, most commonly to the cere­brum, regional lymph nodes, and lung. Squamous cell carcinomas are usually poorly dif­ferentiated and exhibit the same histological fea­tures as squamous cell carcinomas found in the anterior regions of the nasal cavities. The histo­logic features are discussed in detail on page 54. As with adenocarcinomas, squamous cell carcino­mas of the olfactory region frequently invade the brain, but metastases are less common. Diagnostic microscopic criteria for olfactory neu­roblastoma have been defined in detail (Obert et al. 1960) and comprise the following: plexiform intercellular fibrils, poorly defined almost nonex­istent cytoplasm, round to oval nuclei, distinct sharply defined nuclear chromatin, compartmen­talization of sheets of neoplastic cells into lobules by fibrovascular septae, rosettes, and pseudoro­settes. These features are very similar to those ob-

Page 60: Respiratory System

Fig. 75. Adenocarcinoma induced in the ethmoid region of a F344 rat with N-nitrosomethylpiperazine. Widely vary­ing degrees of differentiation are present. Hand E, x 200

served in poorly differentiated adenocarcinomas of the ethmoid region, which undoubtedly has led to diagnostic errors in some cases. For example, electron microscopy and reexamination of histo­logical material from a study in which p-cresidine was reported to induce esthesioneuroblastomas revealed no features of neurogenic origin in the tumors. Origin from basal cells of the olfactory epithelium was suggested and the diagnosis of ad­enocarcinoma was made (Reznik et al. 1981). Di­agnosis of olfactory neuroblastoma should rely on the presence of neurogenic features (best de­monstrated by electron microscopy), such as ax­ons, neurofibrils, neurotubules, and dense-cored secretory granules. Most studies in rats where ol­factory neuroblastomas have been reported have not included this verification, and therefore such diagnoses are in question.

Ultrastructure

Very few studies of tumors in the ethmoid region of the rat nasal cavities have included electron microscopic examination. One neoplasm ob­served ultrastructurally was induced in a F344 rat

Neoplasms, Mucosa, Ethmoid Turbinates, Rat 49

Fig. 76. Squamous differentiation within an adenocarcino­ma induced in the ethmoid region of a F344 rat with N­nitrosomethylpiperazine. Hand E, x 300

by chronic inhalation of the soil fumigant nema­tocide 1,2-dibromo-3-chloropropane (3 ppm, 6 h/ day, 5 days/week). The tumor had infiltrated the brain and nasal bones at the time the animal was killed. By light microscopy, the tumor was classi­fied as poorly differentiated adenocarcinoma. Electron microscopy (Reznik-SchUller, unpub­lished results) revealed junctional complexes which linked the tumor cells together. The cytoplasm contained numerous ribosomes and polyribosomes, while rough endoplasmic ret­iculum and mitochondria were scanty (Fig. 79 and 80). A few of the neoplastic cells possessed long, slender microvilli at their luminal surfaces (Fig. 80). However, in no cell were dense-cored granules, neurotubules or axon-like extensions detected. It was concluded that this tumor was de­rived from basal cells with some differentiation toward sustentacular features.

Differential Diagnosis

The major differential diagnostic problem is be­tween poorly differentiated adenocarcinoma and olfactory neuroblastoma (esthesioneuroblasto-

Page 61: Respiratory System

50 Sherman F. Stinson and Hildegard M. Reznik-Schuller

Fig. 77. Poorly differentiated adenocarcinoma induced in a F344 rat by feeding diets containing p-cresidine for 2 years. Invasion into the bones of the ethmoid region is seen. Hand E, x 220

rna). This problem is compounded by the coexis­tence of epithelial and neurogenic cells in the eth­moid regions of the nasal cavities, as well as by the similarity in appearance of cytological ele­ments and multicellular structures (such as ro­settes) at the light microscopic level. Thus conclu­sive diagnosis without ultrastructural observa­tions is very tenuous. Olfactory rosettes are usually round and are lined by well-differentiated tall columnar cells with bas­ally located nuclei, which do not display marked cytologic atypia (Silva et al. 1983). The rosettes of poorly differentiated adenocarcinomas, on the other hand, are frequently composed of anaplas­tic cells exhibiting considerable pleomorphism (Reznik et al. 1980a, b). However, due to the ab­sence of rosettes in some olfactory neuroblasto­mas (Elkon 1983), differential diagnosis at the light microscopic level should rely on the demon­stration of neurofibrils by a suitable histochemical technique. Definitive differential diagnosis is facilitated by electron microscopic analysis. Ultrastructural fea­tures indicating a neurogenic origin include: in-

Fig.78. Invasion of an adenocarcinoma into the brain from the ethmoid region of a F344 rat fed diets containing p-cresidine for 2 years. Hand E, x 130

tercellular filamentous dendritic processes origi­nating from tumor cell bodies, neurotubules, and intracellular dense-cored secretory granules (EI­kon 1983). When these features are not demon­strated, a diagnosis of adenocarcinoma must be favored over one of neuroblastoma.

Biologic Features

The spontaneous occurrence of neoplasms in the nasal cavities of rats is extremely infrequent. In over 2500 male and female untreated F344 rats held for 2 years, only one benign and one malig­nant neoplasm were found in a single female rat, and these were in the respiratory (anterior) re­gions (Reznik et al. 1980a). No spontaneously oc­curring neoplasms have been reported in the eth­moid regions of the nasal cavities of rats. Treatment of rats with a wide variety of sub­stances by various routes of exposure has been as­sociated with the induction of neoplasms of the ethmoid regions. These have been comprehen­sively reviewed recently (Reznik-Schuller 1983 a; Stinson 1983). Several nitrosamines, given system-

Page 62: Respiratory System

Fig.79. Tumor induced in the olfactory region of a F344 rat by chronic inhalation of 1,2-dibromo-3-chloropropane. The cytoplasm is poorly differentiated, with ribosomes and polyribosomes predominating over other organelles. Electron micrograph, uranyl acetate and lead citrate, x 7000

ically, have been reported to induce neoplasms of the olfactory epithelium. Adenocarcinomas or ol­factory neuroblastomas were diagnosed most fre­quently and squamous cell carcinomas were also reported. Some of the more potent compounds in this respect include N-nitroso-bis (2-hydroxypro­pyl)amine, N-nitrosomorpholine, N-nitroso-2, 6-dimethylmorpholine, N-nitrosopiperidine, N­nitrosomethylpiperazine, N-nitroso-3, 4-dichloro­pyrrolidine, and N-nitrosonornicotine. Other im­portant environmental contaminants also induc­ing tumors of the ethmoid region include: di­bromochloropropane and dibromoethane by in­halation and p-cresidine, procarbazine, 2-me­thoxy-5-methylanaline, and 1-methoxy-4-nitro-te­trachlorobenzene by systemic routes of exposure. Squamous cell carcinomas and adenocarcinomas or olfactory neuroblastomas are frequently in­duced by the same compound and may coexist in the same animal. In a comprehensive serial sacrifice experiment, the pathogenesis of tumors induced in the olfacto­ry region of F344 rats by chronic treatment with

Neoplasms, Mucosa, Ethmoid Turbinates, Rat 51

Fig.SO. Electron micrograph of tumor shown in Fig. 73. Luminal surfaces of tumor cells are lined by long slender microvilli, a marker of sustentacular cells. Uranyl acetate and lead citrate, x 10000

N-nitrosomethylpiperazine (0.13% in drinking water 5 days/week for life) was studied by light and electron microscopy (Reznik-SchUller 1983 b). Mter 4 weeks of treatment, focal hyper­plasias (Fig. 81) developed in the basal layer of the olfactory epithelium. Ultrastructurally, some of these proliferated cells contained dense-cored granules of the neuroendocrine type (Fig. 82), while in others the fine structure of basal cells was evident. Around the 20th week of treatment, some of these lesions were found to have undergone en­dophytic growth, penetrating from the epithelium into the submucosa (Fig.83). From such lesions invasive carcinomas developed, which were com­posed of areas of variable morphological pat­terns. Electron microscopy revealed some cells with neuroendocrine features and others with ele­ments of adenoid or squamous differentiation. In one case, axon-like cytoplasmic extensions indi­cated partial differentiation into neuroblasts. Sim­ilar changes were observed in rats following inha­lation of dibromochloropropane or dibromo­ethane (Reznik et al. 1980 b).

Page 63: Respiratory System

52 Sherman F. Stinson and Hildegard M. Reznik-SchUller

Fig.81 (Above). Olfactory region in a F344 rat treated for 4 weeks with N-nitrosomethylpiperazine. Focal hyperpla­sia of small round to oval cells has developed in the basal epithelial layer. The surface of the lesion is covered by ol­factory sensory and sustentacular cells. Toluidine blue, x 560

Fig. 82 (Below). Basal hyperplasia of cells of olfactory epi­thelium in a F344 rat treated for 4 weeks with N-nitro­somethylpiperazine. Some of the cells contain dense-cored cytoplasmic granules, the ultrastructural marker of neuro­endocrine cells. Electron micrograph, uranyl acetate and lead citrate, x 10200

Page 64: Respiratory System

These observations are consistent with the conclu­sion that neoplasms in the ethmoid region are de­rived from multi potential basal cells in the olfac­tory epithelium, which are capable of various types of differentiation. Taken together with other information discussed previously, these data rein­force the opinion that a diagnosis of olfactory neuroblastoma as a tumor in this region should be made only with conclusive evidence of neurogen­ic origin.

Comparison with Other Species

Neoplasms of the ethmoid regions of the types described have been induced in hamsters, mice, and gerbils. Studies in various species using the same compound and similar dosing regimens sug­gest that the sensitivity of Syrian hamsters to nasal carcinogens is similar to that of rats, while Euro­pean hamsters are more sensitive and Chinese hamsters and mice are less sensitive (Reznik­Schuller 1983 a; Stinson 1983). Few studies have been conducted in other species, but some olfac­tory neoplasms have been reported. Adenocarci­nomas in nonhuman primates and squamous cell carcinomas in dogs have been induced by nitrosa­mines (Reznik-Schuller 1983 a), and adenocarci-

Neoplasms, Mucosa, Ethmoid Turbinates, Rat 53

nomas of the olfactory epithelium have been found in sheep (Young et al. 1961). Eleven nasal tumors classified as esthesioneuroepitheliomas were found among 21600 mammals necropsied at the Philadelphia Zoo (Montali et al. 1983). All were in small carnivores (raccoons, skunks, mink, etc.) and may have been related to housing on contaminated oak planks. As in rats, the spon­taneous occurrence of nasal neoplasms in these species is extremely infrequent. In humans, tumors of the ethmoid regions are rare. Among these, squamous cell carcinomas predominate. Adenocarcinomas and olfactory neuroblastomas, with and without olfactory dif­ferentiation, have also been reported (Heffner 1983). The histological appearance of these tu­mors is similar to that found in rats.

Acknowledgement. This project has been funded in part with Federal funds from the Department of Health and Human Services, under contract number N01-CO-23909 with Litton Bionetics, Inc. The contents of this publication do not neces­sarily reflect the views or policies of the Depart­ment of Health and Human Services; similarly, mention of trade names, commercial products, or organizations does not imply endorsement by the U. S. Government.

Fig.83. Olfactory region of a F344 rat treated for 20 weeks with N-nitrosomethylpiperazine. Proliferating cells from the basal layer of the olfactory epithelium have penetrated the submucosa. Toluidine blue, x 130

Page 65: Respiratory System

54 William D. Kerns

References

Elkon D (1983) Olfactory esthesioneuroblastoma. In: Rez­nik G, Stinson SF (eds) Nasal tumors in animals and man, vol II. Tumor pathology. CRC, Boca Raton, chap 6

Heffner DK (1983) Histopathologic classification of hu­man sino nasal tumors. In: Reznik G, Stinson SF (eds) Nasal tumors in animals and man, vol II. Tumor pathol­ogy. CRC, Boca Raton, chap 1

Montali RJ, Valerio MG, Harshbarger JC (1983) Tumors of the nasal cavity in nondomesticated animals. In: Reznik G, Stinson SF (eds) Nasal tumors in animals and man, vol II. Tumor pathology. CRC, Boca Raton, chapt 11

Obert GJ, Devine KD, McDonald JR (1960) Olfactory neuroblastomas. Cancer 13: 205-215

Reznik G, Reznik-Schuller H, Ward JM, Stinson SF (1980a) Morphology of nasal-cavity tumours in rats af­ter chronic inhalation of 1,2-dibromo-3-chloropropane. Br J Cancer 42: 772-781

Reznik G, Stinson SF, Ward JM (1980b) Respiratory pa­thology in rats and mice after inhalation of 1,2-dibromo-3-chloropropane or 1,2-dibromoethane for 13 weeks. Arch Toxicol46: 233-240

Reznik G, Reznik-Schiiller HM, Hayden DW, Russfield A, Murthy ASK (1981) Morphology of nasal cavity neo­plasms in F344 rats after chronic feeding of p-cresidine, an intermediate of dyes and pigments. Anticancer Res 1 : 279-286

Reznik-Schiiller HM (1983 a) Nitrosamine-induced nasal cavity carcinogenesis. In: Reznik G, Stinson SF (eds)Nasal tumors in animals and man, vol III. Experi­mental nasal carcinogenesis. CRC, Boca Raton, chap 3

Reznik-Schuller HM (1983 b) Pathogenesis of tumors in­duced with N-nitrosomethylpiperazine in the olfactory region of the rat nasal cavity. JNCI 71: 165-172

Silva EG, Mackay B, Butler JJ (1983) Nasal neuroblasto­mas in man. In: Reznik G, Stinson SF (eds) Nasal tu­mors in animals and man, vol II. Tumor pathology. CRC, Boca Raton, chap 4

Stinson SF (1983) Nasal cavity cancer in laboratory animal bioassays of environmental compounds. In: Reznik G, Stinson SF (eds) Nasal tumors in animals and man, vol III. Experimental nasal carcinogenesis. CRC, Boca Ra­ton, chap 7

Young S, Lovelace SA, Hawkins WW Jr, Catlin JE (1961) Neoplasms of the olfactory mucous membrane of sheep. Cornell Vet 51: 96-112

Squamous Cell Carcinoma, Nasal Mucosa, Rat

William D. Kerns

Synonym. Epidermoid carcinoma.

Gross Appearance

Clinically, squamous cell carcinomas, as well as all other malignant neoplasms of the nasal cavity, are invasive tumors that protrude dorsally or lat­erally from the nasal cavity (Fig.84). Frequently, the epidermis overlying the tumor is ulcerated and necrotic. Often, during the early stages of tu­mor growth, a unilateral nasoocular discharge is observed; this can be associated with tumor growth and interference with nasolacrimal drain­age. Once the tumor has penetrated the nasal or maxillary bones, its growth is rapid. Moribund rats exhibit marked dyspnea and emaciation. Macroscopically, squamous cell carcinomas con­tain areas of whitish gray tissue and nests of lami­nated caseous material (keratin). The tumor is usually unilateral and may cause marked com­pression of the contralateral nasal passage (Fig. 85). Squamous cell carcinomas usually origi­nate in the anterior nasal cavity (Kerns et al. 1983;

Lee and Trochimowicz 1982) and frequently ex­tend posteriorly into the ethmoturbinates (Fig. 86) and olfactory bulbs.

Microscopic Features

Squamous cell carcinomas may originate from metaplastic squamous epithelium or from the squamous epithelium that is normally found in the nasal vestibule. The nasoturbinates, maxillo­turbinates, nasal septum, or lateral wall may be af­fected. While invasion of the nasal and maxillary bones or nasal septum is common, tumor growth through the hard palate into the oral cavity is rare. Tumor cell differentiation varies widely and many nasal squamous cell carcinomas are osteolytic. With this type of nasal cancer, as with many oth­ers, poor differentiation is associated with inva­sion of the nasal bone (Lee and Trochimowicz 1982). However, some of the most differentiated carcinomas may also be very invasive (Fig. 87). Excessive keratinization (Fig. 88) and extension of tumor tissue into the ethmoturbinates and naso-

Page 66: Respiratory System

pharyngeal duct are common histomorphological findings, but extension into the brain (Fig. 89) is rare (less than 2%) (Pavkov et al. 1981; Lee and Trochimowicz 1982). When metastases do occur, they are found in the regional lymph nodes and lung. Cytologically, well-differentiated carcinomas are characterized by prominent intracellular bridges, normal keratinization (although abundant in most cases), minimal nuclear atypia, a low mitotic in­dex, and invasion of adjacent structures. Some squamous cell carcinomas are characterized by foci of less differentiated tumor cells (Fig. 90). Al­though keratin (intracellular and extracellular) is normally present in these foci, the tumor cells do not resemble mature squamous cells. They are re­cognized by their nuclear atypia, abnormal kera­tinization (parakeratosis), polygonal and epithe­lioid morphology, abnormal mitotic figures, and absence of intracellular bridges.

Ultrastructure

The cells of well-differentiated squamous carci­nomas are recognized by the presence of tonofila­ments, desmosomes, prominent nucleoli, and tu­bular profiles of rough endoplasmic reticulum (Figs.91 and 92). Cytoplasmic membranes are characterized by elaborate intercellular interdigi­tations. Less differentiated tumors demonstrate a marked decrease in the number of glycogen par­ticulates, desmosomes, tonofilaments, and cell membrane interdigitations (Lee et al. 1983).

Differential Diagnosis

Squamous cell carcinomas should not pose any diagnostic problems. Some less differentiated car­cinomas, especially those with spindle-shaped cells and acinar structures, may require ultrastruc­tural, immunoultrastructural, or immunohisto­chemical (keratin) verification as to the cell of ori­gin (Nagle et al. 1983; N athrath et al. 1982; Wilson et al. 1982).

Biologic Features

With one exception (Lee et al. 1983), squamous cell carcinomas are known to occur only as exper­imentally induced lesions of the nasal cavity in the rat. They have been induced with a variety of related and unrelated compounds by inhalation, as well as by oral, intraperitoneal, and subcutane-

Squamous Cell Carcinoma, Nasal Mucosa, Rat 55

Fig.84. A rat exposed to formaldehyde. The large subcu­taneous mass anterior to the eyes is an invasive squamous cell carcinoma of the nasal mucosa

Fig.85. Coronal section of an undeca1cified head from a rat exposed to formaldehyde. An invasive squamous cell carcinoma protrudes from the nasal cavity and has com­pressed the contralateral nasal passage

Page 67: Respiratory System

56 William D. Kerns

Fig.86. Midsagittal section of a rat exposed to formaldehyde. A destructive squamous cell carcinoma of the anterior nasal cavity extends into the ethmoturbinates (arrow)

Fig. 87. Advanced squamous cell carcinoma that has invaded the nasal bone of a rat exposed to formaldehyde. Hand E, x 180 (reduced by 15%)

ous routes of exposure (Table 3; Stinson 1983). The pathogenesis of this lesion has not been total­ly defined, but necrosis and/or hyperplasia of the respiratory epithelium followed by epidermoid (squamous) metaplasia and dysplasia usually pre­cede neoplasia (Pavkov et al. 1981; Kerns et al. 1983).

Comparison with Other Species

In humans, squamous cell cell carcinoma is the predominant malignant tumor of the anterior na-

sal cavity (Ash et al. 1964). Nuclear pleomor­phism is uncommon and the tumors may show slight or usually no evidence of keratinization. Squamous cell carcinomas in this location rarely metastasize. Workers in the furniture and leather industry are reported to have a higher incidence of nasal cancer (Roush et al. 1980; Andersen et al. 1977; Brinton et al. 1977; Collan 1983; Buiat­ti et al. 1983), and a nasal squamous cell carci­noma has been reported in a patient with a his­tory of formaldehyde exposure (Halperin et al. 1983).

Page 68: Respiratory System

Fig.88. A squamous cell carcinoma, characterized by excessive keratinization, has replaced the naso­turbinate in this rat exposed to formaldehyde. H and E, x 90 (reduced by 15%)

Fig.89. An invasive squamous cell carcinoma has totally replaced the left olfactory bulb in this rat exposed to formaldehyde. Hand E, x 90 (reduced by 15%)

Squamous Cell Carcinoma, Nasal Mucosa, Rat 57

Page 69: Respiratory System

58 William D.Kems

Fig.90 (Above). A poorly differentiated squamous cell car­cinoma of the nasal cavity in a rat exposed to formalde­hyde. Hand E, x 360

Fig.91 (Below). Neoplastic squamous cells from a nasal carcinoma. TEM, x 6000

Page 70: Respiratory System

Squamous Cell Carcinoma, Nasal Mucosa, Rat 59

Table 3. Squamous cell carcinomas observed in the nasal cavity of the rat

Chemical Strain Malea Femalea Study type Reference

Formaldehyde F344 Inhalation Kerns et al. 5.6 ppm 1/119 1/116 1983

14.3 ppm 51/117 52/115

Phenylglycidyl ether S-D Inhalation Lee et al. o ppm 1/89 0/87 1983

12 ppm 9/85 4/89

p-Cresidine F344 2/54 8/47 Feeding Reznik et al. 1.0% 1981

1,2-Dibromo-3-chloropropane F344 Inhalation Reznik et al. 0.6 ppm 4/50 5/50 1980a,b 3.0 ppm 17/45 5/49

Epichlorohydrin S-D Inhalation Laskin et al. 30 ppm 1/100 ND 1980

100 ppm 15/140 ND

Formaldehyde and hydrogen S-D Inhalation Albert et al. chloride 1982

14.7 ppm HCHO and 25/99 ND 10.6 ppm HCl

Hexamethylphosphoramide S-D Inhalation Lee and Trochimowicz 50ppb 24/194b 1982

100ppb 59/200b

400ppb 1371219b

4000ppb 1201215b

Dioxane S-D Drinking Hoch-Ligeti 0.75% 1/30 ND water et al. 1970 1.00% 1/30 ND 1.40% 2/30 ND 1.80% 2/30 ND

Bis( chloromethyl)ether S-D Inhalation Kuschner 0.1 ppm 1/50 ND et al. 1975

l,4-Dioxane Sherman Drinking Kociba et al. 1.0% 1/60 2/60 water 1974

Dimethylcarbamoylchloride S-D Inhalation Sellakumar 1 ppm 96% ND et al. 1980

Phenacetin S-D Feeding Isaka et al. 1.25% 3/22 1/25 1979 2.50% 2127 0127

3,4,5-Trimethoxycinnamaldehyde NR Injection Schoental and Gibbard 150 mg/kg (IP) and 2/4 ND 1972 100 mg/kg (SC)

S-D, Sprague-Dawley; ND, not determined; NR, not reported a No. of tumors/no. of nasal cavities evaluated b Sexes were combined

Page 71: Respiratory System

60 William D.Kerns

In other laboratory animals, chemically induced squamous cell carcinomas have been reported in the mouse (Kerns et al. 1983; Reznik et al. 1980 b), hamster (Sellakumar et al. 1980; Feron et al. 1982), and gerbil (Cardesa et al. 1976). Squamous cell carcinomas have been reported in other spe­cies but are not known to be associated with chemical or infectious agents. In the dog, the males of dolichocephalic breeds are most often affected, and squamous cell carci­nomas were reported to account for 33% of all na­sal and paranasal-sinus carcinomas in one study (Moulton 1978) and 8% of all nasal and parana­sal-sinus neoplasms in another study (Madewell et al. 1976). In the cat, squamous cell carcinoma was the most frequently reported nasal neoplasm, with reported frequencies of 71 % (Moulton 1978) and 44% (Madewell et al. 1976). Squamous cell carcinoma originating in the nasal cavity and sinuses of the horse has also been re­ported (Jean and Daudel 1949; Cotchin 1967), and was found to be the most common nasal neo­plasm (five of 13) in one study (Madewell et al. 1976).

Fig.92. Neoplastic squamous cells from a nasal carcinoma. Note desmosomes (D) and bundles of to no filaments (T). TEM, x 24000 (reduced by 15%)

References

Albert RE, Sellakumar AR, Laskin S, Kuschner M, Nelson N, Snyder CA (1982) Gaseous formaldehyde and hydro­gen chloride induction of nasal cancer in rats. JNCI 68: 597-603

Andersen HC, Andersen I, Solgaard J (1977) Nasal can­cers, symptoms and upper airway function in wood­workers. Br J Ind Med 34: 201-207

Ash JE, Beck MR, Wilkes JD (1964) Atlas of tumor pathol­ogy. Tumors of the upper respiratory tract and ear. Armed Forces Institute of Pathology, Washington, DC, sect IV, fasc 12 and 13

Brinton LA, Blot WJ, Stone BJ, Fraumeni JF Jr (1977) A death certificate analysis of nasal cancer among furni­ture workers in North Carolina. Cancer Res 37: 3473-3474

Buiatti E, Geddes M, Carnevale F, Merler E (1983) Nasal cavity and paranasal sinus tumors in woodworkers and shoemakers in Italy compared to other countries. In: Reznik G, Stinson SF (eds) Nasal tumors in animals and man, vol 1. Anatomy, physiology and epidemiology. CRC, Boca Raton, chap 5

Cardesa A, Pour P, Haas H, Althoff J, Mohr U (1976) His­togenesis of tumors from the nasal cavities induced by diethylnitrosamine. Cancer 37: 346-355

Collan Y (1983) Epidemiologic and etiologic aspects and histopathology of nasal carcinoma in Finland. In:

Page 72: Respiratory System

Reznik G, Stinson SF (eds) Nasal tumors in animals and man, vol 1. Anatomy, physiology and epidemiol­ogy. CRC, Boca Raton, chap 4

Cotchin E (1967) Spontaneous neoplasms of the upper re­spiratory tract in animals. In: Muir CS, Shanmugarat­nam K (eds) Cancer of the naso-pharynx. International Union Against Cancer, Monogr series no 1, Medical Ex­amination, Flushing, pp 203-215

Feron VJ, Kruysse A, Wouters en RA (1982) Respiratory tract tumours in hamsters exposed to acetaldehyde va­pour alone or simultaneously to benzo( a )pyrene or di­ethylnitrosamine. Eur 1 Cancer Clin Oncol18: 13-31

Halperin WE, Goodman M, Stayner L, Elliott LJ, Keenly­side RA, Landrigan Pl (1983) Nasal cancer in a worker exposed to formaldehyde. lAMA 249: 510-512

Hoch-Ligeti C, Argus MF, Arcos lC (1970) Induction of carcinomas in the nasal cavity of rats by dioxane. Br 1 Cancer 24: 164-167

Isaka H, Yoshii H, Otsuji A, Koike M, Nagai Y, Koura M, Sugiyasu K Kanabayashi T (1979) Tumors of Sprague­Dawley rats induced by long-term feeding of phenace­tin. Gan 70: 29-36

Jean R, Daudel R (1949) Epithelioma des sinus et des fosses nasales chez un cheval. Bull Serv Elev Indust Anim AOF 2: 15-21

Kerns WD, Pavkov KL, Donofrio DJ, Gralla EJ, Swen­berg lA (1983) Carcinogenicity of formaldehyde in rats and mice after long-term inhalation exposure. Cancer Res 43: 4382-4392

Kociba RJ, McCollister SB, Park C, Torkelson TR, Geh­ring Pl (1974) 1,4-Dioxane. I. Results of a 2-year inges­tion study in rats. Toxicol Appl Pharmacol 30: 275-286

Kuschner M, Laskin S, Drew RT, Cappiello V, Nelson N (1975) Inhalation carcinogenicity of alpha halo ethers. III. Lifetime and limited period inhalation studies with bis( chloromethyl)ether at 0.1 ppm. Arch Environ Health 30:73-77

Laskin S, Sellakumar AR, Kuschner M, Nelson N, La Mendola S, Rusch GM, Katz GV, Dulak NC, Albert RE (1980) Inhalation carcinogenicity of epichlorohydrin in noninbred Sprague-Dawley rats. JNCI 65: 751-757

Lee KP, Trochimowicz HJ (1982) Induction of nasal tu­mors in rats exposed to hexamethylphosphoramide by inhalation. JNCI 68: 157-171

Lee KP, Schneider PW, Trochimowicz HJ (1983) Morpho­logic expression of glandular differentiation in the epi­dermoid nasal carcinomas induced by phenylglycidyl ether inhalation. Am J Pathol 111: 140-148

Madewell BR, Priester W A, Gillette EL, Snyder SP (1976) Neoplasms of the nasal passages and paranasal sinuses in domesticated animals as reported by 13 veterinary colleges. Am J Vet Res 37: 851-856

Squamous Cell Carcinoma, Nasal Mucosa, Rat 61

Moulton JE (1978) Tumors of the respiratory system. In: Moulton JE (ed) Tumors in domestic animals. Universi­ty of California Press, Berkeley, chap 6

Nagle RB, McDaniel KM, Clark VA, Payne CM (1983) The use of antikeratin antibodies in the diagnosis of hu­man neoplasms. Am J Clin Pathol 79: 458-466

Nathrath WB, Wilson PD, Trejdosiewicz LK (1982) Im­munohistochemical localisation of keratin and luminal epithelial antigen in myoepithelial and luminal epithe­lial cells of human mammary and salivary gland tu­mours. Pathol Res Pract 175: 279-288

Pavkov KL, Kerns WD, Mitchell RI, Connell MM, Do­nofrio DJ, Harroff HH (1981) A chronic inhalation toxi­cology study in rats and mice exposed to formaldehyde. In: Chemical Industry Institute of Toxicology Final Re­port, Dockett no. 10922 Battelle, Columbus Labs, Co­lumbus

Reznik, G, Reznik-Schuller HM, Hayden DW, Russfield A, Murthy ASK (1981) Morphology of nasal cavity neo­plasms in F344 rats after chronic feeding of p-cresidine, an intermediate of dyes and pigments. Anticancer Res 1 : 279-286

Reznik G, Reznik-Schuller HM, Ward 1M, Stinson SF (1980a) Morphology of nasal-cavity tumours in rats af­ter chronic inhalation of 1,2-dibromo-3-chloropropane. Br J Cancer 42: 772-781

Reznik G, Ulland B, Stinson SF, Ward 1M (1980b) Mor­phology and sex-dependent manifestation of nasal tu­mors in B6C3Fl mice after chronic inhalation of 1,2-dibromo-3-chloropropane. 1 Cancer Res Clin Oncol 98:75-83

Roush GC, Meigs lW, Kelly JA, Flannery JT, Burdo H (1980) Sinonasal cancer and occupation: a case-control study. Am 1 Epidemiol111: 183-193

Schoental R, Gibbard S (1972) Nasal and other tumours in rats given 3,4,5,-trimethoxy-cinnamaldehyde, a deriva­tive of sinapaldehyde and other alpha, beta-unsaturated aldehydic wood lignin constituents. Br J Cancer 26: 504-505

Sellakumar AR, Laskin S, Kuschner M, Rusch G, Katz GV, Snyder CA, Albert RE (1980) Inhalation carcino­genesis by dimethy1carbamoyl chloride in Syrian golden hamsters. J Environ Pathol Toxicol4: 107-115

Stinson SF (1983) Nasal cavity cancer in laboratory animal bioassays of environmental compounds. In: Reznik G, Stinson SF (eds) Nasal tumors in animals and man, vol III. Experimental carcinogenesis, CRC, Boca Raton, chap 7

Wilson PD, Nathrath WB, Trejdosiewicz LK (1982) Immu­noelectron microscopic localisation of keratin and lumi­nal epithelial antigens in normal and neoplastic urothe­lium. Pathol Res Pract 175: 289-298

Page 73: Respiratory System

62 Parviz M. Pour

Squamous Cell Carcinoma, Upper Respiratory Tract, Syrian Hamster

Parviz M. Pour

Synonyms. Epidermoid carcinoma; squamous cell tumor.

Gross Appearance

In its earliest stages, a squamous cell carcinoma is not visible grossly. Larger lesions may elevate the mucosa or project from its surface but are not dis­tinctive in any gross features. Invasive tumors ap­pear as grayish white masses penetrating into the surrounding tissue.

Microscopic Features

This neoplasm arises from the epithelium or from the columnar epithelium of ducts or glands just beneath the surface epithelium in the mucosa of the nasal cavity. The initial change in the respira­tory epithelium is disorganization of the epithelial cells followed by a gradual squamous metaplasia. Increasing focal or multifocal atypia of the epi­thelium may reach an extent consistent with carci­noma in situ. The metaplastic epithelium some­times continues to grow and project from the surface to form papillomas. Early squamous cell carcinoma is recognized by the accumulation of many layers of atypical squa­mous epithelial cells which have lost polarity and replaced the respiratory epithelial cells. These squamous cells initially respect the basal laminae, but eventually penetrate them (Figs.93 and 94) and sometimes invade underlying mucous glands. Invasion is evident by small epithelial nests (Fig. 95), which appear as isolated islands beneath the surface epithelium and invade lymphatics or veins (Fig.96). The origin of these malignant ep­ithelial nests cannot always be determined; they may come from the surface epithelium or the sub­mucosal glands.

Ultrastructure

The ultrastructural features of this neoplasm are similar to those of squamous cell carcinomas of the lung in Syrian hamsters (discussed on page 116).

Differential Diagnosis

Most squamous cell carcinomas are well-differen­tiated keratinizing types. However, various de­grees of differentiation are encountered (Figs. 97 and 98). In tumors with a mixture of glandular and squamous epithelium, a diagnosis of muco­epidermoid tumor is justified if both squamous and glandular cells (secreting PAS-positive mate­rial) are present in adequate proportions and in a mixed pattern. Squamous cell carcinoma in the upper respiratory tract must be differentiated from squamous papil­loma and squamous metaplasia (described in de­tail on page 36). Squamous papillomas project from the surface of the respiratory epithelium, re­semble normal stratified squamous epithelium, and do not undergo the loss of polarity, dysplasia, and growth through the basal lamina which are characteristic of squamous cell carcinomas. Hyperplasia may be seen in respiratory epitheli­um with the characteristic increase in number of cells and may accompany squamous metaplasia, in which the pseudostratified columnar cells of the respiratory mucosa are replaced by squamous cells. Papillary projections into the lumen and downgrowth through the basal lamina do not oc­cur. Squamous metaplasia may occur in associa­tion with chronic inflammation and does not ne­cessarily precede neoplasia.

Biologic Features

Natural History. Squamous cell metaplasia of the nasal respiratory epithelium, especially in the an­terior nasal region, is common in several hamster strains (Pour et al. 1976b, 1979). Spontaneous squamous cell tumors are extremely rare. Thus far, in a large number of cases studied, we have seen no carcinomas and only one papilloma in the maxillary turbinate of an albino hamster (Pour 1983), a finding that could indicate either that metaplasia in hamsters does not present a prema­lignant lesion or that the time sequence between metaplasia and malignancy is longer than the usual hamster life span. We tend to believe the first to be true, since metaplasia occurs often dur­ing the inflammatory process.

Page 74: Respiratory System

Squamous Cell Carcinoma, Upper Respiratory Tract, Syrian Hamster 63

Fig. 93 (Above). Early squamous cell carcinoma induced by BHP. Early invasion of the basal membrane (arrows). H and E, x 195 (reduced by 30%)

Most squamous cell carcinomas and mucoepider­moid tumors are invasive initially, rather than ex­pansive (Fig.98). At certain points, the exophytic (expansive) growth begins to invade rapidly, pri­marily in the bones, and these tumors occasional­ly present bulky, fungating masses which extend to the external nares. For some as yet unknown reason, they metastasize infrequently, despite their considerable size. Only rarely do tumors show early invasion of lymphatics and blood ves­sels (Fig.96) and metastasize to the submandibu­lar lymph nodes.

Fig.94 (Below). Dysplastic squamous cell epithelium (bot­tom) and early squamous cell carcinoma invading the sub­mucosa (arrows) in a MPN-treated hamster. Hand E, x 195 (reduced by 30%)

Etiology. Certain nitrosamines are potent inducers of paranasal cavity tumors. Remarkably, the mor­phology of induced lesions can vary from one compound to another. Some of these compounds (Table 1 p.28), such as N-nitrosomethyl(2-oxobu­tyl)amine (M-2-0B) (Pour et al. 1983) and N-ni­trosohexamethyleneimine (N-6-MI) (Althoff et al. 1973), induce adenocarcinomas with a few squa­mous cell tumors. In contrast, N-nitrosobis(2-hy­droxypropyl)amine (BHP) (Pour et al. 1975), N­nitrosodi-n-propylamine (DPN) (Pour et al. 1973, 1974b), N-nitrosomethyl-n-propylamine (MPN)

Page 75: Respiratory System

64 Parviz M. Pour

Fig.95 (Above). Malignant squamous cells (arrows) in the submucosa of the anterior nasal cavity adjacent to hyper­plastic and metaplastic respiratory epithelium. Hand E, x 195 (reduced by 30%)

(Pour et al. 1974 d), 1-acetoxypropylnitrosamine (l-APPN) (Althoff et al. 1977 a), N-nitrosovinyl­ethylamine (VEN) (Althoff et al. 1977b), N-nitro­somethyl(2-oxopropyl)amine (MOP) (Pour et al. 1980), and N-nitrosobis(2-acetoxypropyl)amine (BAP) (Pour et al. 1976a) are potent inducers of squamous cell neoplasms.

Pathogenesis. Squamous cell carcinoma induction was found in most instances to follow squamous cell metaplasia, hyperplasia, dysplasia and in situ

Fig. 96 (Below). Squamous cell carcinoma induced by DPN in the anterior nasal cavity with invasion of lymphatic (straight arrow) and blood vessels (curved arrows). Hand E, x 390 (reduced by 30%)

changes. However, this neoplasm evidently can also develop de novo after high doses of potent carcinogens. The gradual changes in respiratory epithelium can best be demonstrated by serial sac­rifice of animals during the study. The earliest al­terations are characterized by focal proliferation of epithelial cells, which may undergo atypia fol­lowed by squamous cell metaplasia. Whereas metaplastic epithelium sometimes continues to grow and to form small or bulky papillomas, in others increasing focal or multifocal atypia of the

Page 76: Respiratory System

Squamous Cell Carcinoma, Upper Respiratory Tract, Syrian Hamster 65

Fig. 97 (Above). Invasive squamous cell carcinoma induced by 2-0PPN in anterior nasal cavity. No basal membrane could be found around this cellular mass by special stains. Hand E, x 195

Fig.98 (Below). Fairly well-differentiated fungating squa­mous cell carcinoma induced by BHP. Tumor originates from the maxillary turbinate mucosa and invades the sub­mucosa (arrows). Hand E, x 72

Page 77: Respiratory System

66 Parviz M. Pour

epithelium to degrees consistent with carcinoma in situ ensue. At this stage the originally flat epi­thelium becomes exophytic and/or endophytic and (in the latter case) with more or less broad pa­pillae that initially respect the basal laminae. Atypical epithelium often grows into or develops within the submucosal glands and appears inva­sive.

Location. Squamous cell tumors are found pri­marily in the anterior regions, mostly on the naso­turbinates and maxilloturbinates. Squamous cell carcinomas have also occurred in much lower in­cidences in the posterior nasal cavity (Pour et al. 197 4 b), primarily at the floor or ceiling level of the ethmoid region. We have found no tumors that originate in the middle nasal cavity.

Frequency. Although no sexual differences were observed in many of our studies, a male predom­inance was seen in a study using N-6-MI, with a male: female ratio of 2.5: 1 (Althoff et al. 1973). The male preponderance appears to have a signif­icant biologic value, since it correlates with the si­tuation in man and in rats, in which castration in­hibited nasal cancer induction by one of the potent nasal carcinogens (Pour and G6tz 1983). The possible sex hormone dependence of nasal tumors is yet to be determined. The incidence of each histologic tumor type ap­pears to depend on the nature of the carcinogen. A high incidence of squamous cell carcinomas has been seen after BHP (Pour et al. 1975) and MPN treatment (Pour et al. 1974d), whereas DPN (Pour et al. 1973), N-nitroso-2-hydroxypropyl-n­propyl amine (2-HPPN) (Pour et al. 1974c) and N-nitroso-2-oxopropyl-n-propylamine (2-0PPN) (Pour et al. 1974a) primarily induce mucoepider­moid neoplasms in as many as 71 % of the ani­mals. The incidence and mUltiplicity of squamous cell and mucoepidermoid tumors depend on the na­ture of the carcinogen, its dose, duration of treat­ment, and the method employed for histological examination to allow proper recognition of the le­sions (Pour et al. 1976c).

References

Althoff J, Cardesa A, Pour P, Mohr U (1973) Carcinogenic effect of n-nitrosohexamethyleneimine in Syrian golden hamsters. JNCI 50: 323-329

Althoff J, Grandjean C, Pour P, Gold B (1977 a) Local and systemic effects of 1-acetoxypropylnitrosamine in Syrian golden hamsters. Z Krebsforsch 90: 127-140

Althoff J, Grandjean C, Russell L, Pour P (1977b) Vinyl­ethylnitrosamine: a potent respiratory carcinogen in Syrian hamsters. JNCI 58: 439-442

Pour PM (1983) Spontaneous respiratory tract tumors in Syrian hamsters. In: Reznik-Schuller HM (ed) Compar­ative respiratory tract carcinogenesis, vol I. Spontaneous respiratory tract carcinogenesis. CRC, Boca Raton, chap '7

Pour PM, Gotz U (1983) Prevention of N-nitrosobis(2-oxopropyl)amine-induced nasal cavity tumors in rats by orchiectomy. JNCI 70: 353-357

Pour P, KrUger FW, Cardesa A, Althoff J. Mohr U (1973) Carcinogenic effect of di-n-propylnitrosamine in Syrian golden hamsters. JNCI 51: 1019-1027

Pour P, Althoff J, Cardesa A, KrUger F, Mohr U (1974a) Effect of beta-oxidized nitrosamines on Syrian golden hamsters. II. 2-0xopropyl-n-propylnitrosamine. JNCI 52: 1869-1874

Pour P, Cardesa A, Althoff 1. Mohr U (1974b) Tumorige­nesis in the nasal olfactory region of Syrian golden ham­sters as a result of di-n-propylnitrosamine and related compunds. Cancer Res 34: 16-26

Pour P, KrUger FW, Althoff J, Cardesa A, Mohr U (1974c) Effect of beta-oxidized nitrosamines on Syrian golden hamsters. I. 2-Hydroxypropyl-n-propylnitrosamine. JNCI 52: 1245-1249

Pour P, Kruger FW, Cardesa A. Althoff J, Mohr U (1974d) Tumorigenicity of methyl-n-propylnitrosamine in Syrian golden hamsters. JNCI 52: 457-462

Pour P, KrUger FW, Althoff J, Cardesa A, Mohr U (1975) Effect of beta-oxidized nitrosamines on Syrian ham­sters. III. 2,2'-Dihydroxy-di-n-propylnitrosamine. JNCI 54:141-146

Pour P, Althoff J, Gingell R, Kupper R, KrUger F, Mohr U (1976a) N-nitroso-bis(2-acetoxypropyl)amine as a fur­ther pancreatic carcinogen in Syrian golden hamsters. Cancer Res 36: 2877-2884

Pour P, Mohr U, Cardesa A, Althoff J, Kmoch N (1976b) Spontaneous tumors and common diseases in two co­lonies of Syrian hamsters. II. Respiratory tract and di­gestive system. JNCI 56: 937-948

Pour P, Stanton MF, Kuschner M, Laskin S, Shabad LM (1976 c) Tumours of the respiratory tract. In: Turusov VS (ed) Pathology of tumours in laboratory animals, vol 1. Tumours of the rat, part 2. IARC Sci Publ 6: 1-40

Pour P, Althoff J, Salmasi SZ, Stepan K (1979) Spontane­ous tumors and common diseases in three types of ham­sters. JNCI 63: 797-811

Pour P, Gingell R, Langenbach R, Nagel D, Grandjean C, Lawson T, Salmasi S (1980) Carcinogenicity of N-nitro­somethyl(2-oxopropyl)amine in Syrian hamsters. Can­cer Res 40: 3585-3590

Pour PM, Nagel D, Lawson T (1983) Carcinogenicity of N­nitrosomethyl(2-oxobutyl)amine and N-nitrosomethyl­(3-oxobutyl)amine in Syrian hamsters with special refer­ence to the pancreas. Cancer Res 43: 4885-4890

Page 78: Respiratory System

Adenocarcinoma, Anterior Nasal Epithelium, Rat 67

Adenocarcinoma, Anterior Nasal Epithelium, Rat

Sherman F. Stinson and Gerd Reznik

Gross Appearance

In the rat, adenocarcinomas of the upper respira­tory epithelium are found almost exclusively in the nasal cavities, few having been reported in the larynx or trachea. Adenocarcinomas in the anteri­or nasal cavities arise most frequently from the epithelium lining the naso- and maxilloturbinals and the nasal septum (Fig. 99). Derivation from the epithelium lining the lateral walls is less com­mon. The neoplasms appear as pink to gray ses­sile, exophytic masses protruding from the turbi­nals or septum and varying in size from less than a millimeter in diameter to large masses which en-

Fig.99. Cross section through anterior nasal cavities of a F344 rat following inhalation of dibromochloropropane for 106weeks. A large adenocarcinoma originating from the maxilloturbinal (a) and multiple small adenomas of the nasoturbinals (b) and dorsal wall (c) are seen. Hand E, x 5.5

tirely fill the nasal passage, compress the turbi­nals, and displace the nasal septum. Occasionally, adenocarcinomas invade through the bony encasement of the nose and become grossly visible on the external dorsolateral surface as a bulging subcutaneous mass (Fig. 100). As these neoplasms are almost always the result of exposure to nasal carcinogens, their spontaneous occurrence being extremely rare, multiple tumors are not an uncommon finding, and they may also coexist with other nasal tumors, including adeno­mas, papillomas, and squamous cell carcinomas.

Fig. tOO. Cross section through anterior nasal cavities of a F344 rat. Dibromochloropropane inhaled for 106 weeks. A large adenocarcinoma has invaded through the dorsal and lateral walls. Hand E, x 5

Page 79: Respiratory System

68 Sherman F. Stinson and Gerd Reznik

Microscopic Features

A broad and continuous range of degrees of dif­ferentiation, from well-differentiated adenomas and low-grade adenocarcinomas to relatively poorly differentiated highly malignant adenocar­cinomas, can arise from the nasal respiratory epi­thelium. Glandular tumors of the nasal turbinates are usually adenomas and well-differentiated ad­enocarcinomas, while more poorly differentiated adenocarcinomas are found most frequently in the epithelium over the maxilloturbinals, dorsal nasal meatus, and concha. Well-differentiated tumors are formed of cords or sheets of uniform eosinophilic cells with round to oval nuclei, ar­ranged in well-formed glandular and cystic struc­tures (Figs. 101 and 102). These glandular struc­tures are commonly filled with an amorphous, PAS-positive material or cellular debris. The nu­clei have peripherally localized chromatin and, usually, a centrally located nucleus. Mitotic fig­ures are not frequent and are usually normal in

Fig. tOt. Moderately well-differentiated adenocarcinoma (a) of the mucosa of the nasoturbinal in a F 344 rat fed p­cresidine for 2 years. Hand E, x 130

type when found. Invasion is usually minimal in well-differentiated tumors and consists of cords or nests of cells penetrating into the submucosa adjacent to the nasal turbinal or nasal septum and occasionally eroding these structures. The poorly differentiated adenocarcinomas have a more solid appearance and are composed of cells with some cellular and nuclear pleomor­phism (Figs. 103 and 104). Completely to incom­pletely formed acinar structures are surrounded by disorganized sheets and cords of round to elongated cells with widely varying nuclear cyto­plasmic ratios (Fig. 104). A relatively high rate of cell division is evidenced by numerous mitotic figures. Erosion of the bony nasal structures and invasion of the tissues of the nasal well are com­mon and often extensive with large neoplasms (Fig. 100). While most of the nasal adenocarcinomas have a predominant degree of differentiation, areas of well or poorly differentiated architecture can usually be found within a single tumor, making

Fig.t02. Higher magnification of an area from the adeno­carcinoma pictured in Fig. lOt. Hand E, x 330

Page 80: Respiratory System

classification of the degree of differentiation of the neoplasm difficult (Lee et al. 1983).

Ultrastructure

Adenocarcinomas of the nasal respiratory epithe­lium have not been studied by means of electron microscopy in the rat or other laboratory rodents.

Differential Diagnosis

It is difficult to distinguish a well-differentiated adenocarcinoma from an adenoma of the nasal epithelium. Their localization and cell of origin are identical, and the size of the tumor, pattern of growth, and histologic appearance are often very similar. Criteria for malignancy include demon­stration of invasion into the submucosa, bones, or

Fig.103. Papilloma (a) of the nasoturbinal and adenocar­cinoma (b) of the maxilloturbinal in a F 344 rat that inhaled dibromochloropropane for 2 years. Hand E, x 35

Adenocarcinoma, Anterior Nasal Epithelium, Rat 69

cartilage and the presence of traditional cytologic changes, including anaplasia, pleomorphism, and abundant or abnormal mitoses. Another diagnostic problem arises in distinguish­ing between poorly differentiated adenocarcino­mas of the respiratory epithelium and adeno­carcinomas of the olfactory epithelium (see page 47). Both are similar in histologic appear­ance and can be induced by the same compounds. The close spatial relationship of the ethmoid and nasal regions, combined with the infiltrative and expansive behavior of the neoplasms, can create confusion as to the site of origin. Multiple or seri­al sectioning will often give enough information to resolve the problem. Adenocarcinomas of the olfactory epithelium are usually more anaplastic than those of the respiratory epithelium, being composed of cells with scant cytoplasm and very pleomorphic nuclei with large nucleoli. PAS-posi­tive material has not been reported in the aci-

Fig.104. Higher magnification of an area from the adeno­carcinoma in Fig.t03. Note relatively solid pattern of poorly differentiated cells, complete and incomplete for­mation of glands, and a large number of mitotic figures. H and E. x 220

Page 81: Respiratory System

70 Sherman F. Stinson and Gerd Reznik

nar structures of tumors of the olfactory epithe­lium. Ultrastructurally, adenocarcinomas of the olfac­tory epithelium reportedly contain cells showing features of sustentacular cells, such as smooth en­doplasmic reticulum (see page 8). These would not be expected in cells of adenocarcinomas of the respiratory epithelium, although they are yet to be studied with the electron microscope. Final­ly, olfactory adenocarcinomas usually penetrate through the cribriform plate into the brain, while those arising from the respiratory epithelium in­vade laterally through the nasal and maxillary bones. Other tumors of these regions (squamous cell car­cinoma, papilloma, mucoepidermoid tumor, he­mangioma, and hemangiosarcoma) are easily dis­tinguishable on the basis of histologic characteris­tics.

Fig.105. Nasoturbinal of a F344 rat fed p-cresidine for 75 weeks. Note goblet cell hyperplasia and squamous metaplasia of the epithelium and submucosal glands. H and E, x 130

Biologic Features

The spontaneous occurrence of adenocarcinomas of the upper respiratory epithelium in rats is very low. In a series of 1794 male and 1754 female un­treated control F344 rats held for 2 years, only one adenocarcinoma of the trachea was found, and none were found in the larynx or nasal cavities (Goodman et al. 1979). Gross examinations were completed in this study but histologic sections through the nose were not made. In a later report, one adenocarcinoma in the anterior nasal cavity of an untreated control female F344 rat has been described (Reznik et al. 1980a). Several compounds have been associated with the induction of adenocarcinomas of the nasal respi­ratory epithelium; these were comprehensively reviewed recently (Reznik et al. 1981; Reznik­Schuller 1983; Stinson 1983). These compounds include several different nitrosamines and other environmental substances. Nitrosamines have

Fig.106. Nasoturbinal of a F344 rat fed p-cresidine for 75 weeks. Note squamous metaplasia of the epithelium (a) and dysplasia and squamous metaplasia of the submuco­sal glands (b). Hand E, x 330

Page 82: Respiratory System

generally been administered systemically, while other substances have induced nasal adenocarci­nomas by inhalation (dibromochloropropane, di­bromo ethane) or when given in the feed (p-cresi­dine, 2-methoxy-5-methylanaline, phenacetin). Most of these compounds also induce benign na­sal neoplasms (papillomas, papillary adenomas) and other malignant nasal neoplasms (squamous cell carcinoma, poorly differentiated adenocar­cinoma of the ethmoid epithelium). Any of these neoplasms may coexist in the same animal after exposure to these carcinogens. Early morphological events associated with car­cinogenesis in the nasal respiratory epithelium in­clude a reaction of the epithelium typified by dis­orientation of basal and ciliated cells, loss of cilia, and cytomegaly of basal cells, followed by focal hyperplasia, mucous or glandular metaplasia, and squamous metaplasia (Figs. 105 and 106) (Reznik et al. 1980 b). Many of these changes are also ob­served in the submucosal glands of the anterior nasal cavities. The earliest neoplasms observed are papillomas and papillary adenomas. The simi­larity in distribution of the adenomas and adeno­carcinomas, the continuum of histologic features from adenomas to poorly differentiated adeno­carcinomas, and the fact that they are induced by the same compounds make it tempting to specu­late that the adenomas have a malignant poten­tial, although no direct observations have been re­ported to support this hypothesis.

Comparison with Other Species

Adenocarcinomas of the nasal respiratory epithe­lium have been induced in mice and hamsters (Reznik-Schuller 1983; Stinson 1983). These neo­plasms are similar in histologic appearance and distribution to those found in the rat and are in­duced by the same compounds. Syrian hamsters appear to have a sensitivity similar to rats to nasal respiratory carcinogenesis, while European ham­sters are more sensitive and Chinese hamsters and mice less sensitive. These differences, in some cases, may be due to differences in main airstream flow and mucociliary clearance (Schreider 1983). Well-differentiated adenocarcinomas similar to those found in rats have been reported in sheep (Njoku and Chineme 1983). These neoplasms may have an epidemiologic association with afla­toxin-contaminated feed. Nasal respiratory ade­nocarcinomas also occur in dogs (Patniak 1983) with similar geographic and temporal epidemio-

Adenocarcinoma, Anterior Nasal Epithelium, Rat 71

logic patterns to those observed in man (Hayes and Wilson 1983). Adenocarcinomas of nasal respiratory epithelium are found in humans, although this is a rare tumor type (Heffner 1983). The histologic appearance is similar to that described in the rat in many cases. Nasal adenocarcinomas in man are associated with occupational exposure to wood and various metal dusts (chromium, nickel, arsenic) (Buiatti et al. 1983; Torjussen 1983).

References

Buiatti E, Geddes M, Carnevale F, Merler E (1983) Nasal cavity and paranasal sinus tumors in woodworkers and shoemakers in Italy compared to other countries. In: Reznik G, Stinson SF (eds) Nasal tumors in animals and man, vol 1. Anatomy, physiology, and epidemiology. CRC, Boca Raton, chap 5

Goodman DG, Ward JM, Squire RA, Chu KC, Linhart MS (1979) Neoplastic and nonneoplastic lesions in ag­ing F344 rats. Toxicol Appl Pharmacol48: 237-248

Hayes HM Jr, Wilson GP (1983) Comparative aspects of nasal passage carcinoma in dogs with man. In: Reznik G, Stinson SF (eds) Nasal tumors in animals and man, vol 2. Tumor pathology. CRC, Boca Raton, chap 10

Heffner DK (1983) Histopathologic classification of hu­man sinonasal tumors. In: Reznik G, Stinson SF (eds) Nasal tumors in animals and man, vol 2. Tumor patholo­gy. CRC, Boca Raton, chap 1

Lee KP, Schneider PW, Trochimowicz HJ (1983) Morpho­logic expression of glandular differentiation in the epi­dermoid nasal carcinomas induced by phenylglycidyl ether inhalation. Am J Pathol 111: 140-148

Njoku CO, Chineme CN (1983) Neoplasms of the nasal cavity of cattle and sheep. In: Reznik G, Stinson SF (eds) Nasal tumors in animals and man, vol 2. Tumor pathology. CRC, Boca Raton, chap 8

Patniak AK (1983) Canine and feline nasal and paranasal neoplasms: morphology and origin. In: Reznik G, Stin­son SF (eds) Nasal tumors in animals and man, vol 2. Tumor pathology. CRC, Boca Raton, chap 9

Reznik G, Reznik-Schuller H, Ward JM, Stinson SF (1980a) Morphology of nasal-cavity tumours in rats af­ter chronic inhalation of 1,2-dibromo-3-chloropropane. Br J Cancer 42: 772-781

Reznik G, Stinson SF, Ward JM (1980b) Respiratory pa­thology in rats and mice after inhalation of 1,2-dibromo-3-chloropropane or 1,2,dibromoethane for 13 weeks. Arch Toxicol46: 233-240

Reznik G, Reznik-Schuller HM, Hayden DW, Russfield A, Murthy AS (1981) Morphology of nasal cavity neo­plasms in F344 rats after chronic feeding of p-cresidine, an intermediate of dyes and pigments. Anticancer Res 1 : 279-286

Reznik-Schuller HM (1983) Nitrosamine induced nasal cavity carcinogenesis. In: Reznik G, Stinson SF (eds) Nasal tumors in animals and man, vol 3. Experimental nasal carcinogenesis. CRC, Boca Raton, chap 3

Page 83: Respiratory System

72 W. Ellis Giddens Jr. and Roger A. Renne

Schreider JP (1983) Nasal airway anatomy and inhalation deposition in experimental animals and people. In: Reznik G, Stinson SF (eds) Nasal tumors in animals and man, vol 3. Experimental nasal carcinogenesis. eRe, Boca Raton, chap 1

Stinson SF (1983) Nasal cavity cancer in laboratory animal bioassays of environmental compounds. In: Reznik G, Stinson SF (eds) Nasal tumors in animals and man, vol

3. Experimental nasal carcinogenesis. eRe Press, Boca Raton, chap 7

Torjussen W (1983) Nasal cancer in nickel workers. Histo­pathological findings and nickel concentrations in the nasal mucosa of nickel workers, and a short review of chromium and arsenic. In: Reznik G, Stinson SF (eds) Nasal tumors in animals and man, vol 2. Tumor patholo­gy. eRe, Boca Raton, chap 2

Hemangiosarcoma, Nasal Cavity, Mouse

W. Ellis Giddens Jr. and Roger A. Renne

Synonyms. Hemangioendothelioma.

Gross Appearance

The lesion is not usually visible grossly because it arises in the lamina propria of the nasal mucosa. If, however, invasion occurs through the maxilla to the subcutis, a slight focal bulging of the skin results. When the skin is removed, the underlying subcutis is seen to be dark red.

Microscopic Features

Hemangiosarcomas arise from the submucosa of the lateral walls of the nasal cavity. The normal pattern ofloose connective tissue and submucosal glands is disrupted by proliferation of endothelial cells with large vesicular or hyperchromatic nu­clei. Some of these cells contain mitotic figures. They form small vascular channels and sinusoids (Figs. 107 and 108). These neoplastic channels are often filled with blood, giving the tumor a hemor­rhagic appearance. Occasionally, areas of throm­bosis, hemorrhage, and necrosis occur in the neo­plasms in those portions nearer the lumen of the nasal cavity. As the neoplasms expand, erosion and loss of the overlying respiratory epithelium follows, but the principal direction of invasion is lateral toward the turbinate bones, maxillary si­nus, maxilla, marrow space of the maxilla, and the subcutis overlying the maxilla (Fig. 109). These hemangiosarcomas are locally invasive, but there is no evidence of metastasis by way of blood ves­sels or lymphatics. Transplantation and growth of tumor cells within the respiratory tract has not been observed.

Differential Diagnosis

Hemangiosarcomas must be distinguished from hemangiomas and from angiectasis, both of which are also induced by inhalation exposure to propylene oxide. Angiectasis can be distinguished because it is not a neoplasm. The cells are cytolog­ically differentiated and therefore benign. New vascular channels are not formed, but preexisting ones are merely dilated. Hemangiomas are more difficult to distinguish but tend to have larger vas­cular channels than those in hemangiosarcomas. Mitotic figures are fewer or absent and the endo­thelial cells are flattened and have a smaller nu­cleus to cytoplasm ratio. Hemangiomas do not in­vade the maxilla or maxillary space. A complicating factor in inhalation studies is that rhinitis is often present. In the propylene oxide study to be described, inflammation led to the ac­cumulation of serous or purulent exudate in the lumen of the nasal cavity. The nasal mucosa and submucosa were often heavily infiltrated with lymphocytes, neutrophils, histiocytes, and plasma cells. Fibroplasia and congestion were often ob­served.

Biologic Features

Hemangiosarcoma of the nasal cavity as a spon­taneous event is extremely rare in humans (Bomer and Arnold 1971) and animals. Only one report (Rabstein and Peters 1973) of spon­taneous hemangiosarcoma in animals could be found. In this report, only one of 358 BALBI CflCd mice which died or were killed when moribund had a hemangiosarcoma of the eth­moid turbinates.

Page 84: Respiratory System

Fig.t07 (Above). Hemangiosarcoma, submucosa, nasal cavity, mouse. Submucosal glands (G) are disrupted by the proliferation of endothelial cells which form small vascu­lar channels (C). Hemorrhage in nasal cavity (L). A mouse that chronically inhaled 400 ppm propylene oxide. Hand E, x 640 (reduced by 30%)

Experimentally induced hemangiosarcoma in the nasal cavity of mice was observed in a 2-year propylene oxide inhalation study (National Toxi­cology Program 1984; Renne et aI., to be publish­ed). In this study (Table 4),300 B6C3F1 mice were divided into three male and three female groups of 50 each with high-dose (400 ppm), low-dose (200 ppm), and control groups for each sex. Mice were exposed by inhalation 6 h each day, 5 days per week, for 103 weeks.

Hemangiosarcoma, Nasal Cavity, Mouse 73

Fig.t08 (Below). Hemangiosarcoma, submucosa, nasal cavity. Mouse exposed to 400 ppm propylene oxide. Note serous exudate in the lumen (L). Hand E, x 640 (reduced by 30%)

Angiectasis, hemangiomas, and hemangiosarco­mas were found at the end of the study in both male and female mice that received the high dose of 400 ppm propylene oxide (Table 4). All except one of the neoplasms were discovered in appar­ently normal mice that were killed at the end of the study. In this mouse, death occurred as a re­sult of a separate primary neoplasm in another tissue.

Page 85: Respiratory System

74 W. Ellis Giddens Jr. and Roger A. Renne

Fig. 109. Hemangiosarcoma, submucosa of dorsal part of nasal cavity, mouse, following long-term inhalation of 400 ppm propylene oxide. Note the subcutis (S) and invasion ofthe marrow (M) of the maxilla. Hand E, x 160 (reduced by 30%)

Table 4. Nasal lesions in B6C3F1 mice exposed to propylene oxide and in controls'

Male mice Female mice

Oppm 200 ppm 400 ppm o ppm 200 ppm 400 ppm

Angiectasis 0/50 0/50 3/50 0/50 0/50 3/50 Hemangioma 0/50 0/50 5/50 0/50 0/50 3/50 Hemangiosarcoma 0/50 0/50 5/50 0/50 0/50 3/50

• Nasal structure of 50 animals per group was examined microscopically; e.g., five of 50 male high-dose mice had hemangiosarcomas and 13 mice bore lesions

Comparison with Other Species

In the mouse study described, hemangiosarcomas of the nasal cavity were locally invasive, extend­ing into submucosal glands, bone, and bone mar­row of the maxilla, and subcutis around the max­illa. There was no evidence of vascular or lymphatic spread to other tissues. Primary he­mangiosarcomas of the nasal cavity in man, al­though extremely rare, tend to recur locally after excision. They also have a tendency to metasta­size via hematogenous or lymphatic channels to other tissues (Bomer and Arnold 1971).

References

Bomer DS, Arnold GE (1971) Rare tumors of the ear, nose and throat. Acta Otolaryngol [Suppl] (Stockh) 289: 1-25

National Toxicology Program (1984) Inhalation bioassay of propylene oxide for possible carcinogenicity. Termi­nal report. US Department of Health and Human Ser­vices, National Institutes of Health, National Technical Information Service, Springfield, VA

Rabstein LS, Peters RL (1973) Tumors of the kidneys, syn­ovia, exocrine pancreas and nasal cavity in BALB/ cfl

Cd mice. JNCI 51: 999-1006 Renne RA, Giddens WE Jr, Boorman GA, Kovatch R,

Clarke WJ (to be published) Tumors in the nasal cavity of F344 rats and B6CF1 mice induced by inhalation of propylene oxide.

Page 86: Respiratory System

Clear Cell Carcinoma, Larynx, Syrian Hamster 75

Clear Cell Carcinoma, Larynx, Syrian Hamster

Parviz M. Pour

Synonyms. None.

Gross Appearance

Clear cell carcinoma is not usually detected with the naked eye, although the tumor may become large and infiltrate the entire circumference of the larynx (Figs. 110 and 111). In the respiratory tract, the larynx is the usual site, but one lesion has been observed in a male hamster at the bifurcation of the trachea (Pour et al. 1983).

Microscopic Features

The tumor cells have abundant pale, nearly trans­lucent cytoplasm confined within distinct cell bor­ders, giving the cells an irregular polyhedral shape. Nuclei are large and round to ovoid and contain finely distributed chromatin and occa­sionally a large nucleolus (Fig. 112). The cells do not stain with periodic acid-Schiff. They grow in solid sheets and bands and have no particular or­ganization (Fig. 111). Since they invade the sub­mucosal glands from outside without penetrating the lumen, pseudoglandular structures may be seen (Figs. 111 and 112). These tumors appear to arise from the basal layer of the respiratory epithelium or of the submucosal glands. They initially appear as single cells or small colonies within the epithelium and grow by expansion and infiltration into the submucosal stroma, elevating the respiratory epithelium. They tend to infiltrate around the submucosal glands and thereby isolate them from the overlying respi­ratory epithelium (Fig. 112). Metastases to the lungs have been seen (Fig. 113).

Ultrastructure

These lesions have not been studied by electron microscopy.

Differential Diagnosis

Inflammatory lesions with intense proliferation of macrophages must be considered in differential

diagnosis as well as certain types of malignant lymphoma. The exact components of the cyto­plasm of the cells, when determined, should pro­vide good clues as to the nature of the cells in­volved.

Biologic Features

Clear cell carcinomas of the larynx are malignant and have a remarkable potential for invasion and early metastases (Fig. 113). Even small tumors may infiltrate blood vessels. The incidence of these lesions has been reported as 1 %-3% (Pour et al. 1976; Pour 1983). The frequency of precur­sor lesions, which have also been seen in some mutant strains, may be as high as 9% in male ham­sters of different colonies (Pour et al. 1979). In an­imals exposed to certain carcinogens, the frequen­cy of these tumors was not increased, so their development appears to be unrelated to such treatment. Although clear cell carcinoma of the larynx is regarded as a disease of aged hamsters, it has been found in animals only 30 weeks of age. Lesions have been seen more often in males than in females. In an examination of two hamster colonies, the ratio of males to females was 3: 1 (Pour et al. 1976). The etiology of this tumor is un­known.

Comparison with other Other Species

This lesion appears to be limited to the Syrian hamster.

References

Pour PM (1983) Spontaneous respiratory tract tumors in Syrian hamsters. In: Reznik-SchUller HM (ed) Compar­ative respiratory tract carcinogenesis, vol 1. Spontane­ous respiratory tract carcinogenesis. CRC, Boca Raton, chap 7

Pour P, Mohr U, Cardesa A, Althoff J, Kmoch N (1976) Spontaneous tumors and common diseases in two colonies of Syrian hamsters. II. Respiratory tract and digestive system. JNCI 56: 937-948

Pour P, Althoff J, Salmasi SZ, Stepan K (1979) Spontane­ous tumors and common diseases in three types of ham­sters. JNCI 63: 797-811

Page 87: Respiratory System

76 Parviz M. Pour

Fig.HO (Above). Clear cell carcinoma of larynx with infil­tration of the mucosa and submucosa. Hand E, x 26

Fig.Hi (Below). Higher magnification of the same tumor as in Fig. 110, with infiltration of the mucosa and submu­cosa. Remnants of submucosal glands (arrows) and focal sclerosis around tumor cell nests (lower middle portion). H and E. x 80

Page 88: Respiratory System

Fig. 112 (Above). Clear cell carcinoma oflarynx, surround­ing and partially eroding submucosal glands. Hand E, x 390

Clear Cell Carcinoma, Larynx, Syrian Hamster 77

Fig. 113 (Below). Pulmonary metastases of a laryngeal clear cell carcinoma. Hand E, x 195

Page 89: Respiratory System

LESIONS DUE TO INFECTIONS

Murine Respiratory Mycoplasmosis, Upper Respiratory Tract, Rat

Trenton R. Schoeb and 1. Russell Lindsey

Synonyms. Murine chronic respiratory disease; infectious catarrh.

Gross Appearance

Some affected rats have mucopurulent nasal exudate or pink, porphyrin-tinted oculonasal dis­charge, but gross lesions in the upper respiratory tract are, in many cases, not detectable. Exudate can sometimes be found in the nasal passages, trachea, and tympanic cavities. These structures should be disturbed as little as possible during dissection and collection of specimens for culture so as to preserve the quality of the tissues for mi­croscopic examination.

Microscopic Features

The principal lesions of murine respiratory myco­plasmosis in the upper respiratory tract are, in de­creasing order of frequency, rhinitis, otitis media, laryngitis, and tracheitis. All are characterized by: (a) epithelial changes including hypertrophy, hy­perplasia, metaplasia to nonkeratizing squamous or stratified squamous epithelium, and goblet cell hyperplasia; (b) neutrophilic exudation; and (c) accumulation oflymphocytes and plasma cells.

Rhinitis. Normal rat nasal mucosa (Fig. 114) con­tains few lymphocytes except for small numbers around and just anterior to the nasopharynx. In murine respiratory mycoplasmosis, lymphoid cells accumulate diffusely in the subepithelial stroma. Loss of cilia, pseudo glandular epithelial hyperplasia, and goblet cell hyperplasia can be extensive and severe (Fig. liS).

Otitis Media. The middle ears are nearly as fre­quently affected as the nasal passages. The tym­panic cavity may be completely filled with neutro­phils. The lining epithelium, normally simple

squamous or low cuboidal, becomes hyperplastic. Goblet cells are often numerous. In many cases the lumen becomes filled with immature collage­nous connective tissue, leaving only a few glandu­lar spaces containing neutrophils at the boundary representing the original lining (Fig. 116). The cavity may eventually clear but the lining mem­brane remains thickened by connective tissue.

Laryngitis and Tracheitis. The laryngeal submuco­sal glands are in many cases dilated with mucopu­rulent exudate. Epithelial changes are as in other tissues, and the tracheal mucosa can become ex­tremely thickened by epithelial hypertrophy and hyperplasia, with formation of gland-like crypts and severe accumulation of lymphoid cells (Figs. 117 and 118).

Ultrastructure

Mycoplasma pulmonis parasitizes the surface of respiratory epithelial cells (Fig. 119). Various de­generative changes occur, ranging from loss of cilia and vacuolation of cytoplasm to necrosis of scattered individual cells. The mechanisms for these changes are unknown, although damage by the accompanying inflammatory response prob­ably contributes.

Differential Diagnosis

The characteristic respiratory sounds (snuffling) are not consistently present, but in many cases are the only clinical manifestation of the disease. For unknown reasons, in some affected rats porphyrin secretion from the Harderian glands results in ac­cumulation of red material around the eyes and external nares. Some authors have mistakenly identified this pigment as serosanguinous ex­udate. In the natural disease, most rats with these signs are infected with M.pulmonis and one or

Page 90: Respiratory System

Murine Respiratory Mycoplasmosis, Upper Respiratory Tract, Rat 79

Fig.114 (Above). Normal nasal septal mucosa. Hand E, x220

Fig.115 (Below). Nasal septal mucosa in severe murine re­spiratory mycoplasmosis with neutrophilic exudate, flat-

more other agents such as Sendai virus, sialoda­cryoadenitis virus, rat coronavirus, Streptoco~cus pneumoniae, Corynebacterium kutscheri, Bordetel­la bronchiseptica, Klebsiella pneumoniae, Pseu­domonas aeruginosa, Pasteurella pneumotropica, or Streptobacillus moniliformis. Uncomplicated in­fections seldom occur. However, M. pulmonis alone is sufficient to produce the full spectrum of lesions of respiratory mycoplasmosis and no oth­er organism has been shown to produce its char­acteristic lesions in pathogen-free rats. M.pulmo­nis is therefore the primary pathogen, but clearly other agents do modify the course of the natural disease.

tening of the superficial epithelium, loss of cilia, extensive goblet cell hyperplasia with formation of glandular ep­ithelial infoldings, and diffuse accumulation of lympho­cytes and plasma cells. Hand E, x 220

Disease caused by Sendai virus is usually subclin­ical in adult rats and is characterized by necrotiz­ing bronchiolitis (Jacoby et al. 1979). Sialoda­cryoadenitis virus and coronavirus do not cause serious respiratory disease in adult rats and do not appear to be important respiratory pathogens, but they can cause focally necrotizing rhinotracheitis and multifocal interstitial pneumonia (Jacoby et al. 1979). These viral lesions, if found, should not be difficult to differentiate from those of respiratory mycoplasmosis. S. pneumoniae and C. kutscheri have been associated with rhinitis and otitis media, although in most cases M. pul­monis is probably also present. Other bacteria are

Page 91: Respiratory System

80 Trenton R.Schoeb and J.RusseU Lindsey

Fig. 116. Tympanic bulla with purulent exudate in lumen (AJ and fibrosis of the lining membrane. Hand E, x 35

probably little more than opportunistic patho­gens. Diagnosticians must diligently gather all informa­tion necessary to identify all agents present in af­fected rats. Results of histological studies, bacteri­al and mycoplasmal cultures, and serological tests must be carefully considered in making diag­noses. A diagnosis of murine respiratory mycoplasmosis can be supported by enzyme-linked immunosor­bent assay (ELISA) of serum antibodies (Horo­witz and Cassell 1978) and by cultural recovery of the organism. Failure to isolate the organism does not rule out the diagnosis, as the organism can be quite difficult to grow by routine culture methods. Among the difficulties which can be encountered is growth inhibition by certain tissue and medium components (Del Giudice et al. 1980; Kaklamanis et al. 1971; Mardh and Taylor-Robinson 1973; Tauraso 1967). Davidson et al. (1981) have report­ed that cultural isolation and ELISA both detect­ed a high percentage of infected rats, and that combinations of methods increased the detection rate. Culturing multiple sites in the respiratory tract also increased the rate of recovery of organ­isms, but of individual sites the organism was most frequently isolated from the nasopharyngeal duct.

Biologic Features

Several aspects of the natural history of murine respiratory mycoplasmosis need to be clarified. The major mode of transmission is probably via aerosol from affected mothers to neonates, but in utero transmission apparently occurs also. Infec­tion results in a slowly progressive respiratory dis­ease which persists throughout the animal's life. Infected rats can transmit the infection to others but horizontal transmission is slow, even within a cage, and is considerably reduced by increasing the space between cages. Transmission of M.pul­monis via food, water, bedding, and other materi­als has been suggested but not proved. Inasmuch as M. pulmonis has been isolated from wild rats, cotton rats, rabbits, Syrian hamsters, and guinea pigs, these animals could be potential sources of infection. In conventional and experimentally infected rats, the nasal passages and middle ears are the most commonly infected sites; lung lesions are less consistently found. Thus the upper respiratory tract seems to be the source of infection for the distal tract. The extent to which the distal airways and lungs become affected seems to depend on complex interactions among host, organism, and environment. Exposure to ammonia from soiled cage bedding or to purified ammonia increases the severity of upper respiratory lesions and both

Page 92: Respiratory System

Murine Respiratory Mycoplasmosis, Upper Respiratory Tract, Rat 81

Fig. 117 (Above). Normal rat trachea. Hand E, x 330

Fig. 118 (Below). Tracheal mucosa in severe chronic murine respiratory mycoplasmosis, with neutrophilic exudate, ep­ithelial hyperplasia, loss of cilia, and mucosal thickening

the incidence and severity of lung lesions (Broder­son et al. 1976). The mechanisms of this effect re­main unclear, but ammonia greatly increases the growth of M. pulmonis in rat respiratory tracts, particularly in the nasal passages, probably through effects on the host rather than on the or­ganism itself (Schoeb et al. 1982).

with accumulation of many lymphocytes and plasma cells. The dark line at the epithelial surface represents numerous mycoplasmas adherent to the cells (see Fig. 119). Hand E, x 330

Other microbial agents are frequently found in colonies and it is likely that some of them can af­fect the expression of mycoplasmosis. Sendai vi­rus is a likely contributor because, although no such studies in rats have been reported, Sendai vi­rus infection in mice enhances intrapulmonary growth of M.pulmonis (Howard et al. 1978), and

Page 93: Respiratory System

82 Trenton R. Schoeb and 1. Russell Lindsey

Fig. 119. Tracheal epithelial cell with numerous M.pulmonis organisms on its surface. TEM, x 12000

alters functions of alveolar macrophages and inhibits pulmonary bacterial clearance (Jakab 1981). Increased susceptibility is also associated with advancing age, possibly as a result of de­creased immune responsiveness (Cassell et al. 1979). Genetically determined factors are also im­portant inasmuch as LEW rats are more suscepti­ble than F344 rats (Davis and Cassell 1982; Davis et al. 1982). Mycoplasma pulmonis inhabits the surface of cil­iated epithelial cells, as do other mycoplasmas af­fecting the respiratory tract (Cassell et al. 1978). This relationship is undoubtedly fundamental to the initiation and maintenance of infection. For example, it may enable the organism to escape elimination by the mucociliary system, cellular and noncellular inflammatory processes, and spe­cific immune effector mechanisms (Cassell et al. 1978). It seems likely that the nonspecific mito­genic activity of M. pulmonis (Naot et al. 1979) al­ters lymphocyte responsiveness and misdirects or disrupts specific immune responses (Cassell et al. 1979). How M.pulmonis damages epithelial cells is unknown, but it probably competes for essen­tial metabolites or cell components. Production of toxic wastes has been suggested but not proved to be associated with pathogenicity. Mycoplasma pulmonis infection is ubiquitous in conventional rat colonies. Studies have shown by ELISA and cultural isolation that infection is also common in "barrier-maintained" colonies in the United States and Great Britain (Cassell et al.

1981). Rats from these colonies had mild or no le­sions, not the classical lesions described here. The organism has also been found in rats thought to be germ-free (Ganaway et al. 1973).

Comparison with Other Species

With the exception of contagious pleuropneumo­nia (Mycoplasma mycoides) of cattle and goats, which is characterized by fibrinous pleuropneu­monia, most respiratory mycoplasmoses are mor­phologically similar. In mice, lesions of murine re­spiratory mycoplasmosis are similar to those in rats, with a few minor differences. Middle ear fi­brosis like that seen in rats does not occur in mice. Lymphoid accumulations in mice contain a great­er proportion of plasma cells, as do the regional lymph nodes. Syncytial giant cells in the epitheli­um of the nasal passages occur in mice with the disease but not in rats. Lesions of Mycoplasma gallisepticum infection in chickens include chronic suppurative rhinitis, sinusitis, tracheitis and bronchitis with epithelial hypertrophy and hyperplasia, increased mucosal mucus production, and lymphoid cell accumula­tion with follicle formation in the lamina propria. The lesions are thus similar to those of the murine respiratory disease. M. gallisepticum alone usually causes a rather mild upper respiratory disease, but infection is frequently complicated by wild or vac­cine Newcastle disease virus, infectious bronchitis

Page 94: Respiratory System

Murine Respiratory Mycoplasmosis, Upper Respiratory Tract, Rat 83

virus, avian adenovirus, or Escherichia coli, result­ing in more severe disease with extension to the lungs and air sacs. In turkeys, M. gallisepticum produces a disease similar to that in chickens but with even more of a tendency for upper respiratory tract lesions, espe­cially sinusitis, to predominate. Thus the disease is usually called infectious sinusitis. M. meleagri­dis causes a spontaneously resolving air sacculitis also characterized by chronic suppurative inflam­mation, lymphoid infiltration, and epithelial hy­perplasia. Swine infected with Mycoplasma hyopneumoniae develop lesions similar to those of murine respira­tory mycoplasmosis, although bronchiectasis al­most never occurs. Pneumonic lesions with mac­rophage and neutrophil accumulations are more prominent than in the rodent disease, and the gross lesions, discrete gray-red firm masses pre­dominantly in the dependent parts of the lungs, are characteristic of porcine "enzootic pneumo­nia." The natural disease is frequently complicat­ed by other agents such as Pasteurella multocida, Mycoplasma hyorhinis, and swine adenovirus. Lesions of M. pneumoniae infection in humans are not well known because the disease is rarely fatal. However, available descriptions indicate that lesions include peribronchial and perivascu­lar lymphoid infiltrates, acute bronchitis and bronchiolitis, transformation of alveolar epitheli­um to cuboidal type and an alveolar exudate made up chiefly of macrophages. These changes are similar to those of other respiratory mycoplas­moses.

References

Broderson JR, Lindsey JR, Crawford JE (1976) The role of environmental ammonia in respiratory mycoplasmosis of rats. Am J Pathol 85: 115-130

Cassell GH, Davis JI(, Wilborn WH, Wise KS (1978) Pa­thobiology of mycoplasmas. In: Schlessinger D (ed) Mi­crobiology 1978. American Society for Microbiology, Washington DC, pp 399-403

Cassell GH, Lindsey JR, Baker HJ, Davis JK (1979) Myco­plasmal and rickettsial diseases. In: Baker HJ, Lindsey JR, Weisbroth SH (eds) The laboratory rat, volt. Aca­demic, New York, chap 10

Cassell OH, Lindsey JR, Davis JK, Davidson MK, Brown MB, Mayo JO (1981) Detection of natural Mycoplasma pulmonis infection in rats and mice by an enzyme linked immunosorbent assay (ELISA). Lab Anim Sci 31: 676-682

Davidson MK, Lindsey JR, Brown MB, Schoeb TR, Cas­sell GH (1981) Comparison of methods for detection of Mycoplasma pulmonis in experimentally and naturally infected rats. J Clin Microbiol14: 646-655

Davis JI(, Cassell GH (1982) Murine respiratory myco­plasmosis in LEW and F344 rats: strain differences in lesion severity. Vet Pathol19: 280-293

Davis JI(, Thorp RB, Maddox PA, Brown MB, Cassell GH (1982) Murine respiratory mycoplasmosis in F344 and LEW rats: evolution of lesions and lung lymphoid cell populations. Infect Immun 36: 720-729

DeI Giudice RA, Gardella RS, Hopps HE (1980) Cultiva­tion of formerly noncultivable strains of Mycoplasma hyorhinis. CUff Microbiol4: 75-80

Ganaway JR, Allen AM, Moore TD, Bohner HJ (1973) Natural infection of germ-free rats with Mycoplasma pulmonis. J Infect Dis 127: 529-537

Horowitz SA, Cassell GH (1978) Detection of antibodies to Mycoplasma pulmonis by an enzyme linked immuno­sorbent assay. Infect Immun 22: 161-170

Howard CJ, Stott EJ, Taylor G (1978) The effect of pneu­monia induced in mice with Mycoplasma pulmonis on resistance to subsequent bacterial infection and the ef­fect of a respiratory infection with Sendai virus on the resistance of mice to Mycoplasma pulmonis. Gen Micro­bioi 109: 79-87

Jacoby RO, Bhatt PN, Jonas AM (1979) Viral diseases. In: Baker HJ, Lindsey JR, Weisbroth SH (eds) The labora­tory rat, vol 1. Academic, New York, chap 11

Jakab GJ (1981) Interactions between Sendai virus and bacterial pathogens in the murine lung: a review. Lab Anim Sci 31: 170-177

Kaklamanis E, Stavropoulos I(, Thomas L (1971) The my­coplasmacidal action of homogenates of normal tissues. In: Madoff S (ed) Mycoplasma and the L-forms of bac­teria. Gordon and Breach, New York, pp 27-35

Mardh PA, Taylor·Robinson D (1973) New approaches to the isolation of mycoplasmas. Med Mikrobiol Immunol (Berl) 158:259-266

Naot Y, Merchav S, Ben-David E, Ginsburg H (1979) Mitogenic activity of Mycoplasma pulmonis. I. Stimu­lation of rat Band T lymphocytes. Immunology 36: 399-406

Schoeb TR, Davidson MI(, Lindsey JR (1982) Intracage ammonia promotes growth of Mycoplasma pulmonis in the respiratory tract of rats. Infect Immun 38: 212-217

Tauraso NM (1967) Effect of diethylaminoethyl dextran on the growth of mycoplasma in agar. J Bacteriol 93: 1559-1564

Page 95: Respiratory System

84 David G. Brownstein

Sialodacryoadenitis Virus Infection, Upper Respiratory Tract, Rat

David G. Brownstein

Synonyms. Rat submaxillary gland virus infec­tion; SDAV infection.

Gross Appearance

Gross lesions are usually extrarespiratory and limited to mixed or serous salivary glands, exorbi­tal glands, Harderian glands, peri glandular con­nective tissue, cervical lymph nodes, and thymus. The submaxillary and parotid salivary glands are enlarged, pale, and edematous. Intermandibular and cervical connective tissue is gelatinous due to periglandular edema. This edema restricts the ve­nous return in the neck, resulting in distention of the great veins entering the thoracic inlet. Exorbi­tal glands are occasionally enlarged. Harderian glands are swollen and flecked with yellow-gray spots. These foci must be distinguished from nor­mal brown-red mottling of the Harderian gland imparted by its normal content of porphyrin pig-

ment. The cervical lymph nodes are enlarged and the thymus is atrophic. In these cases, ocular le­sions may include corneal opacity, corneal ulcers, pannus, hypopyon, hyphema, and megaloglobus (Innes and Stanton 1961; Jacoby et al. 1975, 1979).

Microscopic Features

Respiratory lesions are primarily restricted to the upper respiratory tract. They precede inflamma­tory changes in the exocrine tissues of the head. Over the course of approximately 5 days, begin­ning on the 2nd day of infection, there is spread­ing necrosis of respiratory epithelium in the nasal cavity accompanied by congestion, edema, and mixed inflammatory infiltrate of the lamina pro­pria (Figs. 120 and 121). The epithelial lining of the turbinates is most severely affected; olfactory epithelium is usually spared. Some meatuses are

Fig. 120. Ventral turbinate and lateral wall of nasal cavity in rat experimentally infected with sialo­dacryoadenitis virus. Note exudative inflammation of the mucosa with gaps in epithelial integrity. H and E, x 240 (reduced by 15%)

Page 96: Respiratory System

Sialodacryoadenitis Virus Infection, Upper Respiratory Tract, Rat 85

covered by exudate composed of necrotic epithe­lium, neutrophils, and mucus. Despite a tropism of this virus for serous or mixed salivary glands, the serous mucosal glands of the nasopharynx sustain relatively mild injury. Necrotic ducts and acini within mucosal glands do occur, however, and afford some specificity to the lesion. There is qualitatively similar inflammation in the trachea, but changes are milder and less uniform than in the nasopharynx. Upper respiratory lesions are resolved by the end of the 2nd week of infection. Lung changes are confined to mild hyperplasia within peribronchial lymphoid nodules (Jacoby et al. 1975, 1979). Severe inflammatory changes occur within mixed or serous salivary glands, exorbital glands, and Harderian glands. Description of these changes is beyond the scope of this volume. The reader is re­ferred to several excellent studies of sequential changes in these tissues (Innes and Stanton 1961; Jacoby et al. 1975, 1979).

Ultrastructure

We have found no report of the ultrastructural features of respiratory tract lesions caused by SDA V, but these features have been studied in in­fected submaxillary gland epithelium (Jonas et al. 1969). Infected epithelial cells have focally dilated cisternae of endoplasmic reticulum and cytoplas­mic vesicles which contain spherical dense or hol­low cores, 60-70 nm in diameter, surrounded by an envelope 80-120 nm in diameter. The charac­teristic corona, seen in negatively stained prepara­tions, is not seen by transmission ultramicroscopy. Morphologically, sialodacryoadenitis virus is in­distinguishable from Parker's rat coronavirus.

Differential Diagnosis

Upper respiratory tract lesions must be distin­guished from those caused by Parker's rat coro­navirus, Sendai virus, pneumonia virus, Myco­plasma pulmonis, and pathogenic bacteria. Pneu­monic changes, which frequently accompany rhinitis caused by Parker's rat coronavirus, Sendai virus, and pneumonia virus, have not been report­ed in SDAV infection. A careful histopathological examination of the exocrine tissues of the head is usually sufficient to enable one to provisionally diagnose SDA V infection, but rhinotracheitis can precede changes in these tissues.

Fig. 121. Ventral turbinate of a rat experimentally infected with sialodacryoadenitis virus. Much of the epithelium is necrotic and desquamated. Some leukocytes are present in the lumen. Hand E, x 740 (reduced by 15%)

Biologic Features

Natural History. Sialodacryoadenitis virus causes acute limited infections; there is no evidence for a carrier state. The virus is highly contagious and is transmitted by aerosol, direct contact, and fo­mites. There are two patterns of infection. Enzo­otic infections occur primarily in breeding colo­nies, where sucklings are passively immune, adults are actively immune, and weanlings are a continuous source of susceptible individuals due to waning passive immunity. It is therefore wean­lings that generally exhibit clinical signs. Explo­sive epizootics occur in non immune colonies with the highest morbidity in weanlings. Signs are usually transient and consist of intermandibular and cervical edema, swelling of submaxillary glands, sneezing, nasal and ocular discharges which are often red-tinged due to a high content of porphyrin, photophobia, and keratoconjuncti­vitis and its sequelae. Some complications of ker­atoconjunctivitis, such as glaucoma and phthisis, cause permanent disfigurement (Jacoby et al.

Page 97: Respiratory System

86 David G. Brownstein

1979). Subclinical infections are common. Exten­sive host range studies have not been done but SDA V can experimentally infect mice by the re­spiratory route (Bhatt et al. 1977).

Pathogenesis. Sialodacryoadenitis virus is epithe­liotropic, with replication limited to the respirato­ry tract and certain exocrine tissues of the head and neck. It replicates at all levels of the respirato­ry tract but produces disease primarily in the upper respiratory tract, where the highest titers are achieved. Virus is excreted for 7 days, after which it is cleared and neutralizing and comple­ment-fixing antibodies appear in the serum (Jaco­by et al. 1975, 1979).

Etiology. Sialodacryoadenitis virus (Coronaviri­dae) is a pleomorphic, enveloped RNA virus with plump. pedunculated surface projections (coro­na). It is approximately 114nm in diameter. The virus replicates intracytoplasmically and virions are formed in cytoplasmic vesicles and endoplas­mic reticulum (Jacoby et al. 1979). The virus is closely related antigenically to Parker's rat coro­navirus (Bhatt et al. 1972).

Frequency. Coronavirus infections are common in commercial and institutional rat colonies (Jacoby et al. 1979; Parker et al. 1970). Because of the close antigenic relationship of SDAV to Parker's rat coronavirus, seroconversion to both viruses occurs in SDAV-infected rats. It is therefore diffi­cult to confirm SDAV infection by serology alone (Bhatt et al. 1972; Jacoby et al. 1979).

Comparison with Other Species

Coronaviruses are ubiquitous in humans, ani­mals, and birds (Bohl 1981). Although coronavi­ruses cause respiratory infections in chickens, hu­mans, and rats, SDAV (and to a limited degree Parker's rat coronavirus) is the only coronarvirus that replicates and produces disease in salivary, exorbital, and Harderian glands.

References

Bhatt PN, Percy DH, Jonas AM (1972) Characterization of the virus of sialodacryoadenitis of rats: a member of the coronavirus group. J Infect Dis 126: 123-130

Bhatt PN, Jacoby RO, Jonas AM (1977) Respiratory infec­tion in mice with sialodacryoadenitis virus, a coronavi­rus of rats. Infect Immun 18: 823-827

Bohl EH (1981) Coronaviruses: diagnosis of infections. In: Kurstak E, Kurstak C (eds) Comparative diagnosis of viral diseases, vol 4. Academic, New York, chap 7

Innes JRM, Stanton MF (1961) Acute diseases of the sub­maxillary and Harderian glands (sialodacryoadenitis) of rats with cytomegaly and no inclusion bodies. Am J Pa­thol 38: 455-468

Jacoby RO, Bhatt PN, Jonas AM (1975) Pathogenesis of sialodacryoadenitis in gnotobiotic rats. Vet Pathol 12: 196-209

Jacoby RO, Bhatt PN, Jonas AM (1979) Viral diseases. In: Baker HJ, Lindsey JR, Weisbroth SH (eds) The labora­tory rat, vol 1. Academic, New York, chapt 11

Jonas AM, Craft J, Black CL, Bhatt PN, Hilding D (1969) Sialodacryoadenitis in the rat. A light and electron mi­croscopic study. Arch Pathol88: 613-622

Parker JC, Cross SS, Rowe WP (1970) Rat coronavirus (RCV): a prevalent, naturally occurring pneumotropic virus of rats. Arch Virusforsch 31: 293-302

Page 98: Respiratory System

The Lung (Bronchi, Bronchioles, Alveolar Ducts, Alveoli, Pleura)

Page 99: Respiratory System

HISTOLOGY AND ULTRASTRUCTURE

Structure and Function of the Lung

Charles Kuhn III

This paper will assume that the outlines of lung structure are known by the reader and will em­phasize some of the newer information that has accumulated in the last few years about the biolo­gy of various types of cells which are found in the lung and airways. It will be convenient to discuss the airways, where bulk flow of air occurs, sepa­rately from the acini, where gas exchange with the circulation takes place.

Conducting Airways

The airways consist of a bifurcating series of mus­cular tubes of considerable complexity. The num­ber ofbranchings varies in different regions of the human lung from eight to perhaps 25 generations before acini are reached. The larger airways are reinforced with cartilage and are called bronchi; airways without cartilage are bronchioles. In the common laboratory rodents, rats, mice, and ham­sters, the only bronchi are extrapulmonary; all airways within the lungs lack cartilage. In large

Fig. 122. Epithelium of rat trachea. Nuclei of the basal cells (a) are darkly stained and oriented horizontally. The pre­dominant columnar cells are ciliated and only scattered

animals, including man, the larger intrapulmo­nary airways also contain cartilage. In the extrapulmonary airways, the cartilage is in the form of a horseshoe-shaped incomplete ring, whereas in intrapulmonary airways of the larger animals it is present as discontinuous islands of cartilage, several at a given level. It is this differ­ence in organization which enables us to cough (Horsfield 1974). When one coughs, one exhales initially against the closed glottis and generates a considerable amount of positive intrathoracic pressure. This pressure would collapse the extra­pulmonary bronchi and trachea if they were not reinforced by the horseshoe-shaped cartilage. The intrapulmonary airways, however, are tethered by the surrounding lung parenchyma so that they will not collapse. However, during the early phase of coughing there is muscular constriction of the bronchi which markedly narrows the lumen. Al­though the discrete islands of cartilage provide some stiffening of the airway walls, they still per­mit them to narrow. Then when the glottis opens, the narrowing of airway lumina results in the very

secretory cells are seen (b). Epoxy section, toluidine blue­basic fuchsin, x 1000

Page 100: Respiratory System

90 Charles Kuhn III

Fig.123. Serous cell of rat bronchial surface epithelium. The secretory granules are electron dense and discrete. Electron micrograph, x 14000

high rate of flow necessary to dislodge material within the airway. The epithelium which lines the airways is a pseu­dostratified ciliated columnar epithelium consist­ing of basal cells, ciliated cells, and secretory cells, all resting on the basal lamina with nuclei at dif­ferent levels (Fig. 122). The basal cells, which do not cause the airway lumen to contract, are the cells which label most heavily with tritiated thy­midine and are presumed to serve as stem cells for the columnar cells. Ciliated and secretory cells are concerned with the production and propulsion of the mucous blanket which provides the mechani­cal protection of the airways against organisms and dusts deposited from the inhaled air. The mucous blanket is conceived as consisting of two phases: a surface layer which consists of mu­cus and an underlying layer of watery fluid in which the cilia beat. The source of watery fluid in this underlying layer is not known with any cer­tainty. No specific cell type has been associated with its production. Its thickness is probably regu­lated by the transport of ions across the airway epithelium, since physiological studies have shown that tracheal epithelium can transport ions

(Nadel et al. 1979). There is a chloride flux from the interstitial space to the bronchial lumen and both a sodium and a chloride flux from the lumen to the interstitium. These ion fluxes will passively carry water osmotically, and this seems a reason­able mechanism to control the thickness of the serous layer. The mucous blanket is a continuous layer. The former controversy about whether it consisted of patches or a continuous coating (van As 1977) was resolved by fixing the tissues by per­fusion through the vascular system, which precip­itates the mucus without dislodging it, revealing the continuous lining (Luchtel1978). The sources of the mucus are twofold - the glands and the surface epithelium. The surface epitheli­um has two types of secretory cells (Jeffery and Reid 1975). The familiar goblet cell has an elec­tron-opaque cytoplasm with a considerable amount of endoplasmic reticulum and has large secretory vesicles with a very pale lucent content. The vesicles are often fused with one another and secretion into the bronchial lumen takes place by compound exocytosis. The other type of secretory cell in rodents is the serous cell of the bronchial surface epithelium (Fig. 123). The cell has a rela­tively electron-lucent cytoplasm but also has an abundance of endoplasmic reticulum. Its secreto­ry granules are discrete and electron dense and do not fuse with one another. The other source of mucus secretion is the mucous glands, which in laboratory rodents such as the hamster or rat are found only in the trachea, but in larger animals such as dog, cat, or man extend as far as the carti­lage does into the peripheral bronchi (Spicer et al. 1971). The mucous glands are compound tubular acinar glands. The more central portions of the tu­bules are lined by cells distended with mucus -the more peripheral parts are lined by serous cells with large apical eosinophilic granules (Fig. 124). By electron microscopy, the distended or mucous cells have relatively electron-lucent secretory vesi­cles and scant, relatively electron-dense cyto­plasm containing endoplasmic reticulum. The ser­ous cell of the tracheobronchial glands is charac­terized by a large amount of lamellar endoplasmic reticulum which occupies the basal portion of the cell. The apical portion of the cell contains O.5-2Ilm discrete electron-dense secretory gra­nules. These granules contain either neutral or acidic glycoproteins. In addition to producing the glycoproteins to be added to the bulk phase of the mucous blanket, serous cells produce specific substances which are probably important in defense against infectious

Page 101: Respiratory System

Fig.124. Seromucous glands from rat trachea. Note large cytoplasmic granules in the serous cells clustered at the ends of secretory tubules (a). On the left, a plasma cells (b)

agents. By immunoperoxidase staining, the gra­nules contain lysozyme (Bowes and Corrin 1977), an enzyme which is active in the hydrolysis of components of certain gram-positive bacterial cell walls, and lactoferrin, an iron-containing protein whose function as an antibacterial agent is not un­derstood. In addition, the serous granules contain a protease inhibitor (Mooren et al. 1982) with a low molecular weight of about 15000 daltons, which inhibits the activity of the leukocyte-de­rived proteases cathepsin G and elastase. As a consequence, this protease inhibitor has been called "antileukoprotease." Its importance lies in the fact that during infection the bronchial mucus becomes purulent and leukocytes degenerate, die, and release potentially injurious proteases. Pre­sumably, the antileukoprotease prevents damage to the cells lining the airways by inactivating the proteases. The third cell in the mucous glands, the myoepi­thelial cell, is represented in the light microscope by elongated nuclei flattened against the basal lamina. By electron microscopy these cells resem­ble smooth muscle; their cytoplasm is filled with contractile filaments with dense attachment bodies. However, the cells lie within the basal lamina and form desmosomal junctions with

Structure and Function of the Lung 91

lies in proximity to the gland. Epoxy section, toluidine blue, x 1000

neighboring mucous and serous secretory cells. Although it is properly an epithelial cell, its ultra­structural features indicate that it is specialized for contraction and allow the inference that its contraction helps to express the mucous secre­tions into the bronchial lumen. Plasma cells are also associated with the mucous glands (Fig. 124). The main immunoglobulin of the mucous secretions is secretory IgA. About 50% of the plasma cells around the mucous glands contain IgA. Both the mucous and serous cells contain a secretory piece in their plasma membrane. The assumption is that the secretory IgA is produced in the plasma cells and is trans­ported across the membrane of secretory cells and into the glandular lumen. The mucous blanket is propelled by the action of the ciliated cells. There are approximately 200 cilia per cell arising from basal bodies in the apex of the cell. Beneath the basal bodies, mitochon­dria are concentrated in the apical cytoplasm to supply the adenosine triphosphate (ATP) re­quired for ciliary beating. The active machinery of the cilia is known as the axoneme (Gibbons 1981). The axoneme consists of nine peripheral doublet pairs of microtubules arranged around the periphery of two central single microtubules

Page 102: Respiratory System

92 Charles Kuhn III

---D

r--~--O

i;;;jjjj ...... ~-R

:.;;;....;;:---- C

(Fig. 125). Attached to each of the doublets at the periphery are a number of accessory structures. Paired arms are attached to one of the tubules of each doublet (the A subfiber). These projecting arms contain an ATPase, dynein, which is in­volved in transduction of the energy from ATP for ciliary beating. Radially arranged structures which extend from the A subfibers toward the central pair are termed "radial spokes." Connect­ing each A subfiber to the B subfiber of the adja­cent doublet is a very fine thread called the "nexin link," which is difficult to see in most electron micrographs. The C-shaped projections attached to the central singlet microtubules are the central sheath. When the cilium beats, it requires magne­sium ions and A TP. In the presence of magnesium the dynein arms form connections to specific sites on the B subfiber of the adjacent doublet. As ATP is hydrolyzed they detach again, but during the detachment they presumably undergo conforma­tional changes and rotate. This rotation produces sliding between the adjacent peripheral doublets (Warner 1978). The sliding is transformed into a bend because the sliding shear is restrained by the presence of the radial spokes which hook up to the central sheath and by the nexin links. This causes the cilium to bend rather than to shoot apart. If trypsin is used to digest the nexin links and the radial spokes, exposure of cilia to A TP causes the cilia to extrude the doublet tubules rather than bend (Lindemann and Gibbons 1975). Why on earth does anybody in veterinary or hu­man medicine want to know so much detail about how cilia work? It turns out that there are subjects

Fig. 125. Cross section of human respiratory cilium. Peripheral microtubular doublet (D), outer dynein arm (0), inner dynein arm (I), radial spoke (R), central singlet microtubule (C). Electron micro­graph, x 200000

in whom cilia fail to beat, producing a condition called the "immotile cilia syndrome" (Mzelius 1981), which occurs in man and has also been de­scribed in the dog. Subjects with immotile cilia have repeated bronchial, sinus, and middle ear in­fections beginning soon after birth. In one partic­ular series from the Sick Children's Hospital in Toronto, about a third of the patients had bron­chiectasis (Corkey et al. 1981). In adults, the pro­portion of patients with bronchiectasis is prob­ably higher. Some patients develop nasal polyps and digital clubbing; males are almost invariably sterile. Half the affected subjects have situs inver­sus. Situs inversus combined with the other features of immotile cilia syndrome is called "Kartagener's syndrome". A variety of structural abnormalities of the cilia give rise to the immotile cilia syn­drome. The most common abnormality is absence of the dynein arms. Other abnormalities which have been described include absence of the radial spokes rather than absence of the dynein arm, se­lective deficiencies of either the outer dynein arm or inner dynein arm alone, and transposition of a microtubular doublet. Within any given family the abnormalities breed true, so presumably there are different genetic abnormalities (Mzelius 1981). Those who are involved in evaluating inhalation toxicology using electron microscopy may come across a variety of abnormal forms of cilia. Many of these are nonspecific abnormalities and do not connote a herditary problem. Chronic irritation to the bronchi can lead to abnormal ciliary struc­tures such as compound cilia, absence of the cen-

Page 103: Respiratory System

Fig. 126. Endocrine-type cells (E) in a neuroepi­thelial body, hamster lung. Cytoplasm is filled with dense core-type granules. Notice unmyelinated nerve (N) beneath the capillary (C). Electron micro­graph, x 12000 (reduced by 15%)

tral tubules or abnormal central tubules, and par­tialloss of dynein arms. Any abnormality which in­volves only a small fraction of the cilia is probably an acquired abnormality and not hereditary. Not all the cells present in the airways can be re­cognized in a hematoxylin and eosin section. Rare cells with well-developed microvilli like those of a brush border (brush cells) can be identified by electron microscopy in airways from trachea to alveolus (Hijiya 1978). Granulated cells can be seen with the aid of silver stains or by using ultra­violet light fluorescence or electron microsco­py (Fig. 126). These cells are called "small gran­ule cells," "endocrine-like cells," or sometimes "Kultschitzky cells" (Bensch et al. 1965). They contain numerous small granules, 0.2-0.4 ~m in diameter, and occur in two forms. Some are dis­crete cells which occur at all levels of the tracheal bronchial tree, in small airways or large airways; in fact, small numbers of them can even be found in the mucous glands. Others are organized cor­puscular collections of argyrophilic cells, which contain both afferent and efferent nerves. The ca­pillaries in the underlying bronchial mucosa asso­ciated with these structures have a fenestrated type of endothelium. These specialized structures have been identified by Lauweryns as neuroepi­thelial bodies.

Structure and Function of the Lung 93

Neuroepithelial bodies occur preferentially near the points of bifurcation of airways. Lauweryns et al. (1977, 1978) have presented evidence that these bodies function as chemoreceptors which are sen­sitive to the oxygen tension in the inspired air. An­imals exposed to hypoxic atmospheres undergo vasoconstriction. Lauweryns' theory is that the stimulation of these organized chemoreceptive structures in the airways mediates hypoxic vaso­constriction. The evidence for this is that cells in the neuroepithelial bodies degranulate when ani­mals are exposed to hypoxia. Within the last few years much has been learned about the contents of those granules. The specific substances identified include serotonin and sever­al peptide hormones identified by immunohisto­chemistry, including calcitonin and the neuropep­tides bombesin and enkephalin (Cutz et al. 1981). The function of these peptides is quite unknown at the present time, but they do serve as potential markers for identifying tumors arising from this cell type. In fact, bombesin seems to be uniformly present in the human bronchial tumor called oat cell carcinoma (Erisman et al. 1982). As one moves from the cartilage-containing-Iayer airways into the bronchioles, there is a shift in the type of epithelium encountered. Ciliated cells still persist but other secretory cells are found: the

Page 104: Respiratory System

94 Charles Kuhn III

Clara cells or nonciliated bronchiolar cells (Fig. 127). The ultrastructural features of the Clara cells are three: they have a conspicuous protrud­ing dome-shaped apex, secretory granules (which in the rat can be either generally spherical or rod­shaped), and a very extensive smooth endoplas­mic reticulum (in most rodents). The granules in Clara cells do not contain mucin. In some species they are PAS positive, in other species they are not. The granules are digestible by pepsin, which is the best available evidence that they contain a basic protein. The granules ap­pear to be secreted by conventional exocytosis to the bronchiolar surface, where they contribute to the bronchiolar lining layer (Kuhn et al. 1974; Ebert et al. 1976). In addition to their secretory function, Clara cells are also important as stem cells in the repair of bronchiolar injury. The ciliated cells are particu­larly susceptible to injury by inhaled toxins, but have little or no capacity to divide, so that the Clara cells are the source of most repair in bron­chiolar injury. One might predict that the Clara cells of rodents, with their extensive smooth endo­plasmic reticulum, would have the enzymatic ma­chinery for metabolizing simple organic chemi-

cals. In fact, Clara cells have even higher levels of cytochrome p4so than do the hepatocytes (Sera­bjit-Singh et al. 1980), and are rather selectively injured by number of simple compounds such as carbon tetrachloride, naphthalene, and 4-ipo­meanol (Boyd et al. 1980). It is uncertain at the present time, however, whether the results of experiments on rodents can be applied to other species, since in many species, including primates, the smooth endoplasmic reti­culum is not well developed. The only endoplas­mic reticulum in the Clara cells in human bron­chioles, for example, is rough endoplasmic reticu­lum. In some rodent species lymphoid nodules scat­tered along the intrapulmonary bronchi are called "bronchus-associated lymphoid tissue," or BALT. BAL T is well developed in rabbits and rats but is absent in the hamster and, probably, in man (McDermott et al. 1982). BALT is a lymphoepi­thelial structure rather similar to the palatine ton­sil. In addition to aggregates of lymphocytes with germinal centers in the submucosa, lymphocytes actually appear in the epithelium of the mucosa. The epithelium overlying the BAL T is nonciliated and seems to be unusually permeable to antigens,

Fig. 127. Bronchiole, rat lung. Brush cell (B). Clara cell (C), ciliated cell (Ci), macrophage (M). Macrophages can be seen on airways as well as alveolar surfaces. Electron micrograph, x 6000

Page 105: Respiratory System

so inhaled antigens may be preferentially trans­ported across this special epithelium to reach the lymphoid tissue. In man there does not seem to be much BAL T, but lympoid nodules do occur near the bifurcations of respiratory and membranous bronchioles and are associated with the beginning of the lymphatic system.

Acini

The gas-exchanging units of the lung are termed "acini" (Weibel 1973). Each acinus is the unit of tissue supplied by a terminal bronchiole and con­sists of respiratory bronchioles, alveolar ducts, alveolar sacs, and their associated alveoli (Fig. 127). In rodents, respiratory bronchioles are almost nonexistent: at most a single short respira­tory bronchiole is present. In man, acini have from two to five and usually three generations of respiratory bronchioles. The pulmonary arteries accompany the bronchioles and enter the acini with the terminal bronchioles (Fig. 127). The arte­rioles accompany the alveolar ducts, giving off ca­pillaries as lateral branches which form a closely woven network in the alveolar walls before drain­ing into the veins at the periphery of the acini. Each vein receives oxygenated blood from two or

Fig.128. Peripheral lung tissue, hamster lung. Bronchiole (B), artery (A), alveolar duct (AD). The artery accompanies the bronchiole and extends along the alveolar duct. The

Structure and Function of the Lung 95

three acini. Thus the arteries accompany the bron­chi while the veins drain separately. Lymphatics start around the small arteries and veins and drain centrally. No lymphatics actually extend out into the alveolar septa. The walls of the distal airspaces have a similar structure whether these are alveoli or alveolar ducts (Fig. 128). They are covered by a thin epithe­lium and consist mainly of capillaries arranged in a grid-like anastomosing pattern best appreciated by focusing up and down on sections much thick­er than the usual diagnostic sections, approxi­mately 50!lm thick. The capillaries are lined by the nonfenestrated type of endothelial cells, joined by tight junctions which are moderately permeable to macromolecules if compared to the epithelium, which has very "tight" junctions (In­oue et al. 1976). The airspace walls are strength­ened by connective tissue bundles containing col­lagenous and elastic fibers. The connective tissue bundles weave back and forth through the capil­lary grid, but in any given section through the air­space wall the connective tissue space lies to one side of the capillary. On the opposite side the ca­pillary is separated from the airspace only by a thin layer of epithelium with which it shares a common basal lamina (Fig. 128). Should fluid fil­tration through the capillaries increase for any

lung was fixed by perfusion at approximately functional residual capacity. SEM, x 100 (reduced by 15%)

Page 106: Respiratory System

96 Charles Kuhn III

Fig.129 (Above). Cross section through interalveolar sep­tum of hamster lung with two adjacent alveoli (A). The cap­illary (C) is separated from the alveolus below by a very de­licate barrier consisting only of attenuated endothelium, epithelium, and shared basal lamina. A type I epithelial cell in the center is sectioned through its nucleus (/). On the left is a portion of a type II cell with lamellar bodies (II). Electron micrograph, x 4000

Fig.130 (Below). Apical portion of a type II alveolar cell from rat lung, with microvilli and characteristic surfactant­containing secretory organelles (lamellar bodies) (arrows). Electron micrograph, x 30000

Page 107: Respiratory System

reason, the fluid will tend to stay within the alveo­lar wall at first because of the tight epithelial junc­tions. The fluid can accumulate in the interstitial space containing connective tissue fibers. How­ever, the capillary remains associated with the alveolar epithelium owing to their shared basal lamina, and considerable edema can accumulate without widening of the diffusion barrier, so that gas exchange is maintained until alveolar flood­ing occurs. The cells of the interstitium are mainly connective tissue cells which differ morphologically from or­dinary fibroblasts in several respects. They have a very irregular outline with complex projections of cytoplasm containing discrete bundles of contrac­tile filaments. The cells contact one another through nexus junctions, so that the contraction of several cells is probably functionally coordi­nated (Bartels 1979). It has been proposed that these cells may have a function in the fine match­ing of ventilation to perfusion (Kapanci et al. 1974). Alveolar epithelium consists of two types of cells. Roughly 98% of the alveolar surface is covered by squamous (type I) epithelial cells from which thin sheets of cytoplasm extend to cover large areas of surface of one or even several alveoli (Haies et al. 1981; Fig. 129). The other (type II) class of ep­ithelial cell is more numerous but smaller, occu­pying only 2% of the alveolar surface. Type II ep­ithelial cells can be recognized in the electron microscope by their characteristic lamellar, phos­pholipid-rich secretory granules, which contain the surface-active alveolar lining material (Askin and Kuhn 1971 ; Fig. 130). In paraffin sections the granules are extracted, and the cells appear vacu­olated. In the nonnallung, few cells are found free in the alveolar spaces, and of those which are present, the great majority (more than 90%) are macro­phages (Hocking and Golde 1979). Most of the lymphocytes are thymus-derived and present in the same relative proportions as in peripheral blood. Macrophages are continuously removed along the airways toward the nares and must be replaced. The majority probably come from circu­lating monocytes (van Oud Alblas et al. 1981), but some come from local sources in the lung intersti­tium (Bowden and Adamson 1980) and perhaps from proliferative pools in the airspaces (Lin et al. 1975). Macrophages produce a variety of secreto­ry products, which are probably important in or­chestrating the inflammatory response and repair of injury (Unanue 1976).

Structure and Function of the Lung 97

References

Mzelius BA (1981) "Immotile-cilia" syndrome and ciliary abnormalities induced by infection and injury. Am Rev Respir Dis 124: 107-109

Askin FB, Kihn C (1971) The cellular origin of pulmonary surfactant. Lab Invest 25: 260-268

Bartels H (1979) Freeze-fracture demonstration of commu­nicating junctions between interstitial cells of the pul­monary interalveolar septa. Am J Anat 155: 125-129

Bensch KG, Gordon GB, Miller LR (1965) Studies on the bronchial counterpart of the Kultschitzky (argentaffin) cell and innervation of bronchial glands. J Ultrastruct Res 12: 668-686

Bowden DH, Adamson IYR (1980) Role of monocytes and interstitial cells in the generation of alveolar macro­phages. I. Kinetic studies of normal mice. Lab Invest 42: 511-517

Bowes D, Corrin B (1977) Ultrastructural immunocyto­chemical localization of lysozyme in human bronchial glands. Thorax 32: 163-170

Boyd MR, Statham CN, Longo NS (1980) The pulmonary Clara cells as a target for toxic chemicals requiring meta­bolic activation. Studies with carbon tetrachloride. J Pharmacol Exp Ther 212: 109-114

Corkey CW, Levison H, Turner JA (1981) The immotile cilia syndrome. A longitudinal survey. Am Rev Respir Dis 124: 544-548

Cutz E, Chan W, Track NS (1981) Bombesin, calcitonin and leu-enkephalin immunoreactivity in endocrine cells of human lung. Experientia 37: 765-767

Ebert RV, Kronenberg RS, Terracio MJ (1976) Study of the surface secretion of the bronchiole using radioautogra­phy. Am Rev Respir Dis 114: 567-573

Erisman MD, Linnoila RI, Hernandez 0, DiAugustine RP, Lazarus LH (1982) Human lung small-cell carcino­ma contains bombesin. Proc Nat! Acad Sci USA 79: 2379-2383

Gibbons IR (1981) Cilia and flagella of eukaryotes. J Cell BioI91:107s-124s

Haies DM, Gil J, Weibel ER (1981) Morphometric study of rat lung cells. I. Numerical and dimensional charac­teristics of parenchymal cell population. Am Rev Respir Dis 123: 533-541

Hijiya K (1978) Electron microscope study of the alveolar brush cell. J Electron Microsc (Tokyo) 27: 223-227

Hocking WG, Golde DW (1979) The pulmonary-alveolar macrophage (Parts I and II). N Engl J Med 301: 580-587,639-645

Horsfield K (1974) The relation between structure and function in the airways of the lung. Br J Dis Chest 68: 145-160

Inoue S, Michel RP, Hogg JC (1976) Zonulae occ1udentes in alveolar epithelium and capillary endothelium of dog lungs studied with the freeze-fracture technique. J UI­trastruct Res 56: 215-225

Jeffery PK, Reid L (1975) New observations of rat airway epithelium: a quantitative and electron microscopic study. J Anat 120: 295-320

Kapanci Y, Assimacopoulos A, Ide C, Zwahlen A, Gab­biani G (1974) "Contractile interstitial cells" in pulmo­nary alveolar septa: a possible regulator of ventilation/ perfusion ratio? Ultrastructural immunofluorescence and in vitro studies. J Cell Bioi 60: 375-392

Page 108: Respiratory System

98 Charles Kuhn III

Kuhn C III, Callaway LA, Askin FB (1974) The forma­tion of granules in the bronchiolar Clara cells of the rat. I. Electron microscopy. J Ultrastruct Res 49 : 387-400

Lauweryns JM, Cokelaere M, Deleersnyder M, Liebens M (1977) Intrapulmonary neuro-epithelial bodies in new­born rabbits. Influence of hypoxia, hyperoxia, hyper­capnea, nicotine, reserpine, L-dopa and 5-HTP. Cell Tis­sue Res 182: 425-440

Lauweryns JM, Cokelaere M, Lerut T, Theunynck P (1978) Cross-circulation studies on the influence of hypoxia and hypoxaemia on neuro-epithelial bodies in young rabbits. Cell Tissue Res 193: 373-386

Lin H-S, Kuhn C, Kuo T-T (1975) Clonal growth of ham­ster free alveolar cells in soft agar. J Exp Med 142: 877-886

Lindemann CB, Gibbons IR (1975) Adenosine triphos­phate-induced motility and sliding of filaments in mam­malian sperm extracted with triton X-l00. J Cell BioI 65 : 147-162

Luchtel DL (1978) The mucous layer of the trachea and major bronchi in the rat. Scan Electron Microsc 2: 1089-1099

McDermott MR, Befus AD, Bienenstock J (1982) The structural basis for immunity in the respiratory tract. Int Rev Exp Pathol23: 47-112

Mooren HWD, Meyer CJLM, Kramps JA, Franken C, Dijkman JH (1982) Ultrastructural localization of the low molecular weight protease inhibitor in human bronchial glands. J Histochem Cytochem 30: 1130-1134

Nadel JA, Davis B, Phipps RJ (1979) Control of mucus se­cretion and ion transport in airways. Annu Rev Physiol 41:369-381

Serabjit-Singh CJ, Wolf CR, Philpot RM, Plopper CG (1980) Cytochrome P-450: localization in rabbit lung. Science 207: 1469-1470

Spicer SS, Chakrin LW, Wardell JR Jr, Kendrick W (1971) Histochemistry of mucosubstances in the canine and human respiratory tract. Lab Invest 25: 483-490

Unanue ER (1976) Secretory function of mononuclear phagocytes. A review. Am J Pathol83: 396-417

van As A (1977) Pulmonary airway clearance mechanisms: a reappraisal. Am Rev Respir Dis 115: 721-726

van Oud Alblas AB, van der Linden-Schrever B, van Furth R (1981) Origin and kinetics of pulmonary macrophages during an inflammatory reaction induced by intrave­nous administration of heat-killed bacillus Calmette­Guerin. J Exp Med 154: 235-252

Warner FD (1978) Cation-induced attachment of ciliary dynein cross-bridges. J Cell BioI 77: R 19-R26

Weibel ER (1973) Morphological basis of alveolar-capil­lary gas exchange. Physiol Rev 53: 419-495

Page 109: Respiratory System

NEOPLASMS

Bronchiolar/Alveolar Adenoma, Lung, Rat

Gary A. Boorman

Synonyms. Pulmonary adenoma: alveologenic adenoma.

Gross Appearance

Spontaneous bronchiolar/alveolar adenoma is usually seen as a solitary spherical gray to white smooth nodule on the pleural or cut surface of the lung. Usually the lesion is 1-5 mm in diameter, sharply demarcated from the surrounding lung parenchyma, and often elevated slightly above the pleural surface.

Microscopic Features

A bronchiolar/alveolar adenoma appears as a fo­cal solid area of increased cellularity that obliter­ates the underlying alveolar architecture (Fig. 131). The lesion is sharply demarcated from the surrounding tissue, with compression and col­lapse of adjacent alveoli (Fig. 133). It contains scant connective tissue stroma. Blood vessels are not a prominent feature and relatively few inflam­matory cells are seen. In contrast to bronchiolar/ alveolar hyperplasia, in which the cells appear to grow along existing alveolar septa, cells of bron­chiolar/alveolar adenoma form solid, glandular or papillary patterns with obliteration of underly­ing alveolar structures. In the glandular pattern the cells are cuboidal to tall columnar, enclose central lumina (Fig. 132), and have moderate cel­lular atypia. Mitotic figures are commonly found. The papillary pattern appears similar, with the cells covering a fibrovascular core and forming papillary projections. In some tumors or areas of tumors the cells grow in a solid pattern (Fig. 134) and do not form linear rows. The cells are fairly uniform, with a moderate amount of cytoplasm and poorly defined boundaries. As opposed to the glandular pattern, in which tumor cell nuclei are often oblong, cells in solid areas have round

nuclei of variable size with mild atypia and some mitotic figures. In hematoxylin and eosin sections the cells are usually more basophilic. Important features for distinguishing bronchiolar/alveolar adenoma are distinct borders, compression of ad­jacent tissue, and obliteration of the underlying architecture.

Ultrastructure

Chemically induced bronchiolar/alveolar adeno­mas in F 344 rats are characterized by the pres­ence of osmiophilic lamellated organelles, sug­gesting that the cells are of type II pneumocyte origin (Reznik-Schuller and Reznik 1982). Cells with ultrastructural features of Clara cells, ciliated cells, mucous cells, or APUD-type cells were not found. Ultrastructural studies of the rare spon­taneous bronchiolar/alveolar adenomas have not appeared in the literature.

Differential Diagnosis

Bronchiolar/alveolar adenoma must be differen­tiated from bronchiolar/alveolar hyperplasia and bronchiolar/alveolar carcinoma. The adenoma appears morphologically to be a stage in progres­sion from hyperplasia to carcinoma. Thus some confusion exists in distinguishing the benign stage. One might argue that since this process ap­pears to represent a spectrum of lesions progres­sing to carcinoma, all lesions, even the earliest ones, might logically be called carcinomas. How­ever, it has not been demonstrated that all lesions progress to carcinoma, and some may even disap­pear when the stimulus is removed. Therefore, it seems prudent to separate lesions into hyperpla­sia. adenomas, and carcinomas using standard morphological criteria that have been shown to be relevant for other species and organ systems until we have additional information regarding the bio-

Page 110: Respiratory System

100 Gary A. Boorman

Fig.131 (Upper left). Bronchiolar/alveolar adenoma, lung, rat. A well-circumscribed lesion with a glandular pattern. Hand E, x 70

Fig. 132 (Lower left). Bronchiolar/alveolar adenoma with a glandular pattern. The nuclei form serpentine rows and the cells tend to surround lumina. Hand E, x 300

Fig. 133 (Upper right). Bronchiolar/alveolar adenoma that is sharply demarcated from surrounding parenchyma. There is collapse and compression of the adjacent alveoli. Hand E, x 100

Fig. 134 (Lower right). Bronchiolar/alveolar adenoma with a solid pattern. The cells are round and have moderate cytoplasm, poorly defined cell boundaries, and few mitotic figures. The underlying alveolar architecture is not discern­ible. Hand E, x 240

Page 111: Respiratory System

logic behavior of these lesions. Certainly, a chemi­cal that induces carcinomas with obvious malig­nant characteristics seems to pose a greater risk to rats than a chemical that after 2 years' exposure only induces lesions that might be called adeno­mas. Thus separation of these lesions into classi­cal categories may allow a more reasoned judg­ment of potential hazard of the chemical and certainly will be useful retrospectively as more be­comes known about the biologic behavior of these lesions. Bronchiolar/alveolar adenoma can be distinguished from hyperplasia by its sharp de­marcation from surrounding parenchyma, loss of underlying alveolar septal architecture and, in some cases, greater cellular atypia. Adenomas tend to undergo a regular pattern of growth, do not stimulate a scirrhous reaction, and do not have the obvious malignant characteristics such as invasion or distant metastases found in carci­nomas.

Biologic Features

Bronchiolar/alveolar adenomas are uncommon and were found in only 18 of 2379 (0.8%) female F344 rats and 35 of 2320 (1.5%) male F344 rats in recent 2-year NTP/NCI studies (Haseman et al. 1984). Four adenomas were found in 365 male and one adenoma in 365 female Sprague-Dawley rats in studies terminated at about 2 years (Stula 1975). In the NCI/NTP 2-year toxicology studies, several chemicals were suggested to cause pul­monary tumors in rats (Table 5). In this extensive testing program rats were exposed by gavage, in­halation, dosed feed, dosed water, or skin paint­ing to approximately 300 chemicals, of which 150 were found to be carcinogenic in one or more sex and species. It is interesting to note (Table 5) that in only four cases were the results judged to be positive on the basis of the lung as a target tissue.

Bronchiolarl Alveolar Adenoma, Lung, Rat 101

Table 5. Chemically induced lung tumors in F 344 rats

Chemical Sex Dose Adeno- Carcino-group masb masb

5-Nitroacenaph- M Control 0/96 1/96 (1) thene Low 2/38 (5) 5/38 (13)

High 3/45 (7) 0/45

5-Nitroacenaph- F Control 1/99 (1) 0/99 thene Low 4/46 (9) 4/46 (9)

High 2/31 (6) 1/31 (3)

2,4,5-Trimethyl- F Control 0120 0129 aniline Low 1/43 (2) 2/43 (5)

High 9/50 (18) 2/50 (4)

1,2-Dibromo- F Control 0/50 0/50 ethane" Low 0/48 0/48

High 1/47 (2) 4/47 (9)

" 1,2-Dibromoethane was an inhalation study, the rest of the chemicals were given in the feed. Number of rats with tumors/rats with lung examined

b Number of rats with tumor/rats with lung examined (0/0)

Another important factor would seem to be that only recently have chemicals been given by inha­lation. Spontaneous bronchiolar/alveolar adeno­mas are very uncommon and little is known about their biologic behavior.

References

Haseman JK, Huff JE, Boorman GA (1984) Use of his tori­cal control data in carcinogenicity studies in rodents. Toxicol Pathol (in press)

Reznik-Schuller HM, Reznik G (1982) Morphology of spontaneous and induced tumors in the bronchiolo­alveolar region of F344 rats. Anticancer Res 2: 53-57

Stula EF (1975) Naturally occurring pulmonary tumors of epithelial origin in Charles-River CD rats. Bull Soc Pharm Environ Pathol3: 3-11

Page 112: Respiratory System

102 Shirley L. Kauffman and Tamiko Sato

Alveolar Type II Cell Adenoma, Lung, Mouse

Shirley L. Kauffman and Tamiko Sato

Synonyms. Type II cell adenoma; pulmonary adenoma; bronchiolo-alveolar adenoma; alveo­logenic adenoma.

Gross Appearance

Alveolar adenomas are pearly white, well demar­cated spherical tumors, visible beneath the pleural surface or protruding from it. The majority mea­sure 2-4 mm in diameter but some reach 1.0 cm. They are typically firm to rubbery, solid, and homogeneous.

Microscopic Features

Type II adenomas consist of a homogeneous population of uniform-sized cuboidal cells re­sembling normal type II respiratory epithelium (Figs.135 and 136). They may be situated in any part of the lung parenchyma but are most fre­quent in the periphery, often near the pleural sur­face. Individual cells are 121lm with central spherical, hyperchromatic nuclei measuring 5-71lm in diameter. The cytoplasm is eosinophilic in formalin-fixed, paraffin-embedded tissues stained with hematoxylin and eosin, and the char-

acteristic lamellar bodies are represented by fine vacuoles (Fig. 137). The latter stain black in glutaraldehyde-fixed postosmicated tissues em­bedded in plastic (Fig. 138). The histological pat­tern is generally trabecular with anastomosing cords forming solid nests; glands or tubules are uncommon except in old tumors. Cell cords are separated on one side by basal alveolar capillaries and sparse reticulin fibers, while the opposite lu­minal border faces a residual airspace. These un­encapsulated tumors have irregular borders creat­ed by the extension of neoplastic cells along adjacent alveolar walls. Macrophages containing ingested lipid and inclusions are frequent along the periphery.

Ultrastructure

Type II adenoma cells resemble normal alveolar cells in that they contain characteristic lamellar bodies, large mitochondria (Fig. 139), and promi­nent Golgi zones (Fig. 140). Tubular nuclear inclu­sions have been described in both murine and hu­man tumors (Flaks and Flaks 1970; Torikata and Ishiwata 1977): Both type A and type C particles have been demonstrated in carcinogen-induced

Fig. 135. Type II adenoma, lung, mouse. Note discrete cords of type II cells lining alveolar walls. Hand E, x 60

Page 113: Respiratory System

Fig. 136. Type II adenoma with homogeneous cell popula-[> tion, trabecular pattern, and irregular but well-demarcated border. Hand E, x 60

'V Fig.137 (Left) Higher magnification of tumor in Fig.136. Note the large spherical nuclei and abundant eosinophilic, vacuolated cytoplasm typical of type II adenoma cells. H and E, x 500

Fig. 138 (Right). Type II adenoma, lung, mouse. Character­istic lamellar bodies seen at the light microscopic level in tissue fixed in glutaraldehyde and postosmicated, x 500

Alveolar Type II Cell Adenoma, Lung, Mouse 103

Page 114: Respiratory System

104 Shirley L. Kauffman and Tamiko Sato

Fig.139 (Left). Cells of type II adenoma, lung, mouse. Note large mitochondria and lamellar bodies with parallel membrane stacks. Microvilli line the central portion of the luminal surface; the remainder is covered by flattened sur­face epithelium. Glutaraldehyde, postosmicated, TEM, x 12000 (reduced by 15%)

type II adenomas (Bucciarelli and Ribacchi 1972) and in carcinomas arising from them (Kimura et al. 1972). Terminal bars are present and cell junc­tions are relatively straight. The luminal surfaces bear central microvilli, which are covered laterally by cytoplasmic flaps of adjacent type I cells, as in normal lung (Brooks 1968). By scanning electron microscopy (Sato and Kauffman 1980) the tumors are seen to consist of nodular masses, generally spherical, with relatively smooth outer surfaces (Fig. 141). The smooth lining is interrupted by protrusions of the microvillus cap of type II cells through the surrounding type I cytoplasm (Fig. 142).

Differential Diagnosis

Type II adenomas should be distinguished from hyperplasia due to toxic injury or viral infection,

Fig. 140 (Right). Apex of a type II adenoma cell with prom­inent Golgi zone, smooth endoplasmic reticulum, and free ribosomes. TEM, x 12000 (reduced by 15%)

adenomatosis (Hom et al. 1952), and Clara cell adenomas. Distinguishing features of type II ad­enomas are trabecular pattern, homogeneous cell population, uniform round nuclei and, ultrastruc­turally, their characteristic lamellar bodies, large mitochondria, and straight plasma membranes. Surfactant-specific apoprotein has been detected in both the nucleus and cytoplasm of human type II adenoma cells (Singh et al. 1981).

Biologic Features

Descriptions of the natural history of mouse lung adenomas frequently include both type II and Clara cell adenomas, as these have only recently been distinguished. Experimentally induced tu­mors are recognizable days to weeks after carci­nogen exposure, and may be preceded by alveolar epithelial hyperplasia (Shimkin 1955; Kauffman 1976). In the early stages, tumors consist of dis-

Page 115: Respiratory System

Fig.141 (Left). Type II cell adenoma, lung, mouse. Smooth-surfaced lobules of type II cells seen protruding from alveoli and alveolar ducts. SEM, x. 450 (reduced by 15%)

crete cords of cuboidal cells lining alveoli (Grady and Stewart 1940) (Fig. 135). Characteristic solid trabecular tumors form as the adjacent affected alveoli collapse. Initial growth is rapid, and vol­ume-doubling time of tumors 1-2 months of age has been estimated as 4.4 days (Shimkin and Po­lissar 1955). With aging, growth rate progressively decreases, due in part to a decrease in the growth fraction (Dyson and Heppleston 1976). Malig­nant tumors have been described arising both spontaneously and after carcinogen exposure. These are presumed to arise in preexisting ade­nomas (see bronchiolar adenomas). Studies of Amaral-Mendes (1969) indicated that approxi­mately 19% of spontaneous alveolar tumors in mice 2 years and older were malignant. Metas­tases are rare (1%-4% of all lung tumors); most frequent sites are local lymph nodes, heart, and diaphragm (Stewart et al. 1979; Turosov et al. 1974). Established cell lines derived from type II tumors have maintained their differentiated ul­trastructural features and secretion of pulmonary surfactant (Stoner et al. 1975).

Alveolar Type II Cell Adenoma, Lung, Mouse 105

Fig. 142 (Right). Type II cell adenoma, mouse. A single tu­mor lobule with discrete protrusions of microvillus caps through the ruffled to flattened epithelium of type I cells. SEM, x 4500 (reduced by 15%)

Etiology and Frequency

Adenomas have been induced by a variety of car­cinogens: chemical and physical agents, food ad­ditives, and environmental dusts and fumes (Shimkin 1955; Shimkin and Stoner 1975). Data on the frequency of spontaneous lung adenomas (both alveolar and Clara cell) in several mouse strains is shown in Table 6. For all lung adenomas genetic constitution is the main determinant, and frequency ranges from 70% in the most suscepti­ble A strain to 1 % in resistant C 57 BL. Suscepti­bility of the same strains to carcinogen-induced adenomas parallels natural frequency within the same strain and appears to be regulated by the major histocompatability complex, H2 (Demant and Cleton 1980). The difference in frequency of gross adenomas induced by urethane in sensitive versus relatively resistant strains may. be due to differences in a single gene (Malkinson and Beer 1983). Evaluation of the histologic types of adeno­mas induced in C 57 strains has demonstrated an association of particular H2 haplotypes with

Page 116: Respiratory System

106 Shirley L. Kauffman and Tamiko Sato

Table 6. Frequency of pulmonary tumors in eight strains of mice (modified from Shimkin 1955)

Strain

A Swiss BALB c(C) I Y C3H dba CS7 leaden (L or M) CS7 black

Pulmonary tumors per 100 animals 12-18 months old

70-90 40-50 15-25 10-20 10-20 5-15 5 1 1

the development of alveolar type II adenomas (Oomen et al. 1983).

Comparison with Other Species

In man, neoplasms arising from type II alveolar cells are classed as a subgroup of pulmonary ad­enocarcinomas, termed "bronchioloalveolar car­cinoma." These are distinguished from the more common terminal bronchiolar carcinomas by their histology, typical osmiophilic lamellar inclu­sions (Bonikos et al. 1977), and surfactant-specific apoprotein (Singh et al. 1981). Familial cases of alveolar cell carcinoma, with and without coexis­tent interstitial pulmonary fibrosis, have been de­scribed (Beaumont et al. 1981; Joishy et al. 1977). Although the peripheral bronchioloalveolar carci­nomas in man are frequently associated with scarred parenchyma and have been termed "scar cancers," no such association has been shown in spontaneous or induced mouse tumors. Adeno­mas resembling those of mice occur in a variety of birds and animals (Stewart 1966), and the simi­larity of jaagsiekte, sheep pulmonary carcinoma, to both type II and Clara cell carcinoma of man has been noted (see Clara cell adenoma).

References

Amaral-Mendes JJ (1969) Histopathology of primary lung tumours in the mouse. J Pathol97: 415-427

Beaumont F, Jansen HM, Elema JD, ten Kate LP, Sluiter HJ (1981) Simultaneous occurrence of pulmonary inter­stitial fibrosis and alveolar cell carcinoma in one family. Thorax 36: 252-258

Bonikos DS, Hendrickson M, Bensch KG (1977) Pulmo­nary alveolar cell carcinoma. Fine structural and in vitro study of a case and critical review of this entity. Am J Surg Pathol1: 93-108

Brooks RE (1968) Pulmonary adenoma of strain A mice: an electron microscopic study. JNCI 41: 719-742

Bucciarelli E, Ribacchi R (1972) C-type particles in pri­mary and transplanted lung tumors induced in BALBI c mice by hydrazine sulfate: electron microscopic and im­munodiffusion studies. JNCI 49: 673-684

Demant P, Cleton FJ (1980) Histocompatibility genes and neoplasia. In: Cleton FJ, Simons JW (eds) Genetic ori­gins of tumor cells. Nijhoff, The Hague, pp 109-125

Dyson P, Heppleston AG (1976) Cell kinetics of urethane­induced murine pulmonary adenomata. II. The growth fraction and cell loss factor. Br J Cancer 33: 105-111

Flaks B, Flaks A (1970) Fine structure of nuclear inclu-sions in murine pulmonary tumor cells. Cancer Res 30: 1437-1443

Grady HG, Stewart HL (1940) Histogenesis of induced pulmonary tumors in strain A mice. Am J Pathol 16: 417-432 + 3 plates

Horn HA, Congdon CC, Eschenbrenner AB, Andervont HB, Stewart HL (1952) Pulmonary adenomatosis in mice. JNCI 12: 1297-1315

Joishy SK, Cooper RA, Rowley PT (1977) Alveolar cell carcinoma in identical twins. Similarity in time of onset, histochemistry, and site of metastasis. Ann Intern Med 87:447-450

Kauffman SL (1976) Autoradiographic study of type II­cell hyperplasia in lungs of mice chronically exposed to urethane. Cell Tissue Kinet 9: 489-497

Kimura I, Miyake T, Ishimoto A, Ito Y (1972) Intracister­nal A-type and C-type particles observed in pulmonary tumors in mice. Gan 63: 563-573

Malkinson AM, Beer DS (1983) Major effect on suscepti­bility to urethan-induced pulmonary adenoma by a single gene in BALB/cBY mice. JNCI 70: 931-936

Oomen LCJM, Demant P, Hart AAM, Emmelot P (1983) Multiple genes in the H-2 complex affect differently the number and growth rate of transplacentally induced lung tumours in mice. Int J Cancer 31: 447 -454

Sato T, Kauffman SL (1980) A scanning electron micro­scopic study of the type 2 and Clara cell adenoma of the mouse lung. Lab Invest 43: 28-36

Shimkin MB (1955) Pulmonary tumors in experimental animals. Adv Cancer Res 3: 223-267

Shimkin MB, Polissar MJ (1955) Some quantitative obser­vations on the induction and growth of primary pul­monary tumors in strain A mice receiving urethan. JNCI 16:75-97

Shimkin MH, Stoner GD (1975) Lung tumors in mice: ap­plication to carcinogenesis bioassay. Adv Cancer Res 21: 1-58

Page 117: Respiratory System

Singh G, Katyal SL, Torikata C (1981) Carcinoma of type 2 pneumocytes. Immunodiagnosis of a subtype of "bron­chioloalveolar carcinomas." Am J Pathol102: 195-208

Stewart" HL (1966) Comparison of histologic lung cancer types in captive wild mammals and birds and laboratory and domestic animals In: Severi L (ed) Lung tumours in animals. Div of Cancer Res, Perugia, pp 25-58

Stewart HL, Dunn TB, Snell KC, Deringer MK (1979) Tu­mours of the respiratory tract. In: Turusov VS (ed) Pa­thology of tumours in laboratory animals, vol II, Tu­mours of the mouse. IARC Sci Pub123: 251-287

Bronchiolar Adenoma, Lung, Mouse

Shirley L. Kauffman and Tamiko Sato

Synonyms. Clara cell adenoma; papillary adeno­ma; bronchioloalveolar adenoma.

Gross Appearance:

These adenomas are pearly white, spherical, sub­pleural tumors 2-4 mm in diameter, indistinguish­able grossly from type II adenomas. Larger tu­mors, 0.5-1.0 cm in diameter, are frequently tan. Multiple mulberry-shaped nodules may protrude from the cut surfaces. Adenomas arise in the pleu­ral, subpleural, and deeper parenchyma of the lung, and are equally represented in different lobes of the lung.

Microscopic Features

Neoplastic Clara cells are cuboidal to columnar, resembling the nonciliated terminal bronchiolar epithelium of normal mouse lung. The nuclei vary in size and in shape from spherical to elongated, and frequently exhibit deep nuclear folds and in­vaginations. Whereas the cytoplasm appears pale in paraffin-embedded, formalin-fixed tissues stained with hematoxylin and eosin, abundant small vacuoles and osmiophilic particles are de­monstrated in tumors fixed by glutaraldehyde and postosmicated (Fig. 143). Clara cell adeno­mas grow in either a tubular or papillary pattern. In the former, cuboidal or columnar cells are en­closed in cylindrical tubes separated by connec­tive tissue septa (Fig. 143). Macrophages, cell se­cretion, and debris frequently fill the tubular or

Alveolar Type II Cell Adenoma, Lung, Mouse 107

Stoner GD, Kikkawa Y, Kniazeff AJ, Miyai K, Wagner RM (1975) Clonal isolation of epithelial cells from mouse lung adenoma. Cancer Res 35: 2177-2185

Torikata C, Ishiwata K (1977) Intranuclear tubular struc­tures observed in the cells of an alveolar cell carcinoma of the lung. Cancer 40: 1194-1201

Turusov VS, Breslow NE, Tomatis L (1974) Frequency and organ distribution of lung tumor metastases in CF-l mice. JNCI 52: 225-232

Fig. 143. Tubular Clara cell adenoma. Columnar cells with irregular, elongated nuclei line well-formed tubules. Mac­rophages, cell debris, and secretions are present in the lu­men. Numerous small cytoplasmic vacuoles and osmio­philic particles are seen. Glutaraldehyde, postosmicated, Epon-embedded, x 500

Page 118: Respiratory System

108 Shirley L. Kauffman and Tamiko Sato

Fig. 144. Tubular Clara cell adenoma with several papillary stalks. Secretory activity of the lining cells is evident. Hand E, x 60

acinar spaces. Large tubular tumors may have pa­pillary areas (Fig. 144). Typical mature papillary tumors consist of elongated vascular stalks lined by cuboidal to tall columnar cells with variable amounts of sequestrated secretions (Fig. 145). Clara cell adenomas have no true capsule but their margins are usually clearly outlined by a rim of compressed lung tissue.

Ultrastructure

Striking ultrastructural features of Clara cell ad­enomas (Kauffman et al. 1979) include nuclear in­foldings and complex interdigitations with adja­cent cells (Figs.146 and 147). Mitochondria are small in comparison to those of type II adenoma cells, and while these may have ill-defined cristae with dark matrices similar to Clara cells of normal mouse, the matrices are often pale and the cristae prominent (Fig. 146). Endoplasmic reticulum is abundant, glycogen may be present, and secretion granules measuring 300-500 nm and membrane­enclosed crystals are characteristic (Fig. 147). My-

Fig. 145. A typical mature papillary adenoma in which vas­cular stalks, containing dense collagen, are lined by cu­boidal to columnar epithelium. Hand E, x 250

elin figures resembling those in injured Clara cells have been described in neoplastic Clara cells (Kennedy et al. 1977). By scanning electron microscopy (Sato and Kauffman 1980) Clara cell tumor surfaces present lobules consisting of closely apposed epithelial cells separated by slightly depressed furrows creating grape-like clusters (Fig.148). Microvilli cover the entire cell surface except for a central apical zone, which is smooth to slightly ruffled (Fig. 149).

Differential Diagnosis

Clara cell adenoma should be distinguished from type II alveolar adenoma and adenomatosis, the latter a benign condition in which mucin-contain­ing columnar epithelial cells line alveolar septa (Hom et al. 1952). Mouse Clara cells are mucin negative. Differential features suggesting Clara cell origin include papillary or tubular pattern, he­terogeneity of nuclear size and shape, and charac­teristic nuclear folds. By electron microscopy, typ-

Page 119: Respiratory System

Fig. 146. Electron micrograph of columnar epithelial cells of a papillary adenoma with microvilli along the luminal surfaces, small dense bodies, mitochondria with pale ma­trices, and abundant endoplasmic reticulum. x 6000

ical features are 300-500 nm secretory granules, crystals, glycogen granules, nuclear invaginations, and complex interdigitating plasma membranes.

Biologic Features

As noted under alveolar adenoma, the natural his­tories of the two histologic types of adenomas have been treated together in the past. More infor­mation is now becoming available regarding the pathogenesis of Clara cell tumors. Clara cell ad­enomas appear to arise either in the peripheral air sacs or form hyperplastic foci in bronchioles and alveolar ducts (Shami et al. 1982). Kennedy et al. (1977) described bronchiolization of alveoli fol­lowed by nodules of hyperplastic Clara cells as precursors of the carcinomas induced in hamsters by polonium-210. Bronchiolar adenomas seldom or never arise within bronchi, although large tu­mors do protrude into them. With age, merging of adjacent adenomas may convert an entire lung lobe into a multilobulated tumor mass.

Bronchiolar Adenoma, Lung, Mouse 109

Fig. 147. Tubular Clara cell adenoma with irregular infold­ings of the nuclear membranes, complex interdigitations of plasma membranes, dense bodies, and membrane-en­closed crystals. x 12000

The latent period is variable, depending upon strain, age, dose, type of carcinogen and route of exposure. Following transplacental carcinogen exposure, tubular clara cell tumors have been re­cognized in Swiss mice at 10 days of age and pa­pillary tumors after 22 days. Studies of the subse­quent progression of these tumors indicated that the papillary tumors arose from an early tubular form (Kauffman 1981). In nude mice, latent peri­ods of up to 9 months have been found for papil­lary tumors (Anderson 1978). The delayed ap­pearance of papillary compared to alveolar tu­mors has been noted by several investigators (Grady and Stewart 1940; Kimura 1971). The lat­ter investigator hypothesized a progression from benign to malignant tumors in which alveolar ad­enomas became papillary and then carcinoma­tous. Amaral-Mendes, on the other hand, classified ad­enomas as either of alveolar or bronchiolar origin and found that the latter gave rise to tumors of greater malignancy (Amaral-Mendes 1969). Me­tastases have been described in 1 %-4% of both

Page 120: Respiratory System

110 Shirley L. Kauffman and Tamiko Sato

Fig. 148. Scanning electron microscopic view of a papillary adenoma in which large grape-like clusters of Clara cells protrude into an airspace. x 450

spontaneous and induced tumors (Stewart et al. 1979; Turusov et al. 1974). Malignant cell lines have been derived from ethyl-nitrosourea-in­duced Clara cell tumors, which grow both as sub­cutaneous papillary and ascites tumors when transplanted to nude mice (Parsa and Kauffman 1983).

Etiology and Frequency

Available data on the etiology (Shimkin 1955; Shimkin and Stoner 1975) and on the frequency of lung adenomas (see alveolar adenoma, Table 6) have not distinguished these according to cell of origin. However, there have been some studies which deal specifically with induction of Clara cell tumors. Adenomas induced in the tree shrew treated with 2,2' -dihydroxy-di-n-propylnitrosa­mine (Rao and Reddy 1980) and the peripheral tumors arising in hamsters exposed to 210pO were all of Clara cell origin (Kennedy etal. 1977). In

Fig.149. Surfaces of Clara cell clusters covered by micro­villi except for smooth central atypical regions. x 4500

the mouse, Clara cell tumors comprised approxi­mately 50% of the total adenomas induced by transplacental exposure to ethyl nitrosourea (Kauffman 1981).

Comparison with Other Species

Clara cell carcinomas were first recognized in man (Montes et al. 1966) and comprise one type of bronchioloalveolar tumors of the peripheral adenocarcinoma group (WHO 1981). These grow along the alveolar septa, are frequently papillary, and are mucin positive. Despite some attempts to establish the origin of human bronchioloalveolar carcinomas from one cell type (either type II or Clara cells), current studies indicate the likeli­hood that there are two distinct tumors. Clara cell tumors have been distinguished from type II car­cinomas ultrastructurally by their characteristic nuclear profiles and cytoplasmic granules (Green­berg et al. 1975), and granules of glycoprotein similar to those of normal Clara cell have been

Page 121: Respiratory System

demonstrated by autoradiography in Clara cell tumors (Dermer 1982). Surfactant-specific apo­protein detected in type II tumors has not been demonstrated in several Clara cell tumors (Singh et al. 1981). The naturally occurring pulmonary carcinoma of sheep (jaagsiekte) has ultrastructur­al features resembling both Clara cell and type II carcinoma and has been proposed as a model of the disease in man (Nobel and Perk 1978). In ad­dition to sheep, domesticated animals with a rela­tively high incidence of peripheral adenocarcino­ma include dogs, cats, horses, and cattle (Balo 1966). In dogs, three-quarters of all primary lung tumors are said to be bronchiolar carcinomas; these are frequently subpleural, may be multicen­tric, are acinar or papillary with mucin produc­tion, and may metastasize (Nielsen 1966).

References

Amaral-Mendes JJ (1969) Histopathology of primary lung tumours in the mouse. J Pathol97: 415-427

Anderson LM (1978) Two crops of primary lung tumors in BALBI c mice after a single transplacental exposure to urethane. Cancer Lett 5: 55-59

Bala J (1966) Comparison of lung tumours in animals and man. In: Severi L (ed) Lung tumours in animals. Div Cancer Res, Perugia, pp 127-139

Dermer GB (1982) Origin of bronchioloalveolar carcino­ma and peripheral bronchial adenocarcinoma. Cancer 49:881-887

Grady HG, Stewart HL (1940) Histogenesis of induced pulmonary tumors in strain A mice. Am J Pathol 16: 417-432 + 3 plates

Greenberg SD, Smith MN, Spjut HJ (1975) Bronchiolo­alveolar carcinoma - cell of origin. Am J Clin Pathol63: 153-167

Hom HA, Congdon CC, Eschenbrenner AB, Andervont HB, Stewart HL (1952) Pulmonary adenomatosis in mice. JNCI 12: 1297-1315

Kauffman SL (1981) Histogenesis of the papillary Clara cell adenoma. Am J Pathol103: 174-180

Kauffman SL, Alexander L, Sass L (1979) Histologic and ultrastructural features of the Clara cell adenoma of the mouse lung. Lab Invest 40: 708-716

Bronchiolar Adenoma, Lung, Mouse 111

Kennedy AR, McGandy RB, Little JB (1977) Histochemi­cal, light and electron microscopic study of polonium-210 induced peripheral tumors in hamster lungs: evi­dence implicating the Clara cell as the cell of origin. Eur J Cancer 13: 1325-1340

Kimura I (1971) Progression of pulmonary tumor in mice. I. Histological studies of primary and transplanted pul­monary tumors. Acta Pathol Jpn 21: 13-56

Montes M, Adler RH, Brennan JC (1966) Bronchiolar apocrine tumor. Am Rev Respir Dis 93: 946-950

Nielsen SW (1966) Spontaneous canine pulmonary tu­mors. In: Severi L (ed) Lung tumours in animals. Div Cancer Res, Perugia, pp 151-164

Nobel TA, Perk K (1978) Bronchiolo-alveolar cell carcino­ma. Animal model: pulmonary adenomatosis of sheep, pulmonary carcinoma of sheep (jaagsiekte). Am J Pa­thol90: 783-786

Pars a I, Kauffman SL (1983) Malignant Clara cell line de­rived from ethyl-nitrosourea-induced murine lung ade­nomas. Cancer Lett 18: 311-316

Rao MS, Reddy JK (1980) Carcinogenicity of 2,2'-dihy­droxy-di-n-propylnitrosamine in the tree shrew (Tupaia glis): light and electron microscopic features of pulmo­nary adenomas. JNCI 65: 835-840

Sato T, Kauffman SL (1980) A scanning electron micro­scopic study of the type 2 and Clara cell adenoma of the mouse lung. Lab Invest 43: 28-36

Shami SG, Thibodeau LA, Kennedy AR, Little JB (1982) Proliferative and morphological changes in the pulmo­nary epithelium of the Syrian golden hamster during carcinogenesis initiated by 21oPoa-radiation. Cancer Res 42: 1405-1411

Shimkin MB (1955) Pulmonary tumors in experimental animals. Adv Cancer Res 3: 223-267

Shimkin MB, Stoner GD (1975) Lung tumors in mice: ap­plication to carcinogenesis bioassay. Adv Cancer Res 21:1-58

Singh, G, Katyal SL, Torikata C (1981) Carcinoma of type 2 pneumocytes. Immunodiagnosis of a subtype of "bronchioloalveolar carcinomas". Am J Pathol 102: 195-208

Stewart HL, Dunn TB, Snell KC, Deringer MK (1979) Tu­mours of the respiratory tract. In: Turusov VS (ed) Pathology of tumours in laboratory animals, vol II. Tumours of the mouse. IARC Sci Pub123: 251-287

Turusov VS, Breslow NE, Tomatis L (1974) Frequency and organ distribution of lung tumor metastases in CF-1 mice. JNCI 52: 225-232

WHO (1981) Histological typing of lung tumours. Tumori 67: 253-272

Page 122: Respiratory System

112 Gary A. Boorman

Bronchiolar/ Alveolar Carcinoma, Lung, Rat

Gary A. Boorman

Synonyms. Bronchiolar carcinoma; alveolar cell carcinoma; adenocarcinoma; carcinoma.

Gross Appearance

While small tumors may not be visible to the un­aided eye, most bronchiolar/alveolar carcinomas are of sufficient size to be easily seen. They ap­pear as firm, pale, white to pink irregular smooth nodules projecting above the pleural surface. In advanced cases an entire lobe may be replaced by tumorous tissue. While in man they are most often found in the upper lobe, a site propensity has not been reported for the rat. When cut most will-have a smooth surface, except for those containing areas of necrosis, which will appear grossly as yel­low to pale brown irregular areas of soft caseous material within the tumor.

Microscopic Features

Spontaneous bronchiolar/alveolar carcinomas are usually solitary tumors that tend to occur in the peripheral regions of the lung (Fig. 150). The tumors often are not well circumscribed and in­vade pleura, vessels, or adjacent bronchi and bronchioles (Fig. 151). The tumor cells may grow along existing alveolar structures or form solid, papillary, and glandular patterns which obliterate the underlying alveolar architecture. The cells are round (in solid patterns) or columnar (papillary or glandular patterns) (Fig. 152), with moderate amounts of cytoplasm and poorly defined cell boundaries. In less differentiated tumors the cells are more pleomorphic and spindle-shaped. These tumors appear to incite a scirrhous response. In some tumors, areas of squamous metaplasia may be found (Fig. 153).

Ultrastructure

Spontaneous and induced bronchiolar/alveolar tumors in the F344 rat are characterized by the presence of intracytoplasmic bodies with cross­barred parallel lamellae analogous to those found in type II pneumocytes (Reznik-SchUller and Rez­nik 1982).

Fig.iSO. Bronchiolar/alveolar carcinoma obliterating nor­mal alveolar architecture. Hand E, x 120

Differential Diagnosis

Bronchiolar/alveolar carcinoma must be differ­entiated from bronchiolar/alveolar adenoma, me­tastatic adenocarcinoma, and squamous cell car­cinoma. Bronchiolar/alveolar adenomas tend to be better differentiated and lack obvious malig­nant characteristics such as distant metastases or local invasion. Size is important, since lesions greater than 1 cm in diameter can usually be shown to have areas suggestive of local invasion. Metastatic carcinomas from the reproductive tract or gastrointestinal tract or of bile duct origin can be mistaken for bronchiolar/alveolar carcino­ma. Metastatic carcinomas are often multiple, tend to occur in lymphatics or blood vessels, and have a more discrete margin. Usually, at the edge of a bronchiolar/alveolar carcinoma the tumor cells can be found growing along existing alveolar

Page 123: Respiratory System

Bronchiolar/Alveolar Carcinoma, Lung, Rat 113

Fig.1S1. Cells of bronchiolar/alveolar carcinoma invading wall of large bronchus (L, lumen of bronchus; M, smooth muscle wall of bronchus). Hand E, x 120

Fig.1S2. Cells of bronchiolar/ alveolar carcinoma forming a glandular pattern. The cells surrounding lumina tend to be columnar. Hand E, x 400

Fig.1S3. Bronchiolar/alveolar carcinoma with area of squamous metaplasia (arrowhead). Hand E, x 200

Page 124: Respiratory System

114 Gary A. Boorman

structures, while in metastatic tumors a sharper demarcation exists between tumor cells and nor­mal pulmonary tissue. Abundant mucus produc­tion is rare in bronchiolar/alveolar carcinomas, but may be a prominent feature in many adeno­carcinomas that can metastasize to the lung. A careful search for a primary tumor in other organs or tissues is a necessary prerequisite before ac­cepting the diagnosis of primary bronchiolar/ alveolar carcinoma. Finally, this tumor must be differentiated from squamous cell carcinoma. In man, carcinomas showing features of both squamous cell carcino­ma and adenocarcinoma behave as adenocar­cinoma (WHO 1982). In the rat, tumors compris­ing a mixture of squamous cells and bronchio­lar/alveolar cells should be diagnosed as bron­chiolar/alveolar carcinoma. The diagnosis of squamous cell carcinoma should be reserved for tumors composed entirely of squamous cells and felt to be of bronchogenic origin. Squamous cell carcinomas meeting these criteria are rare in the rat, but when found should be more central in location than bronchiolar/alveolar carcinomas, with local areas of squamous differentiation.

Biologic Features

Morphologically, bronchiolar/alveolar carcino­ma appears to represent a progression of the bronchiolar/alveolar adenoma. The exact biolog­ic behavior has not been established, and whether these lesions are malignant from their inception or undergo malignant changes with time has not been established. The limited ultrastructural studies (Reznik-Schuller et al. 1981; Reznik­Schuller and Reznik 1982) suggest that some, if not most, of these tumors arise from type Ilpneu­mocytes. In man, bronchiolar/alveolar carcino­mas are placed in a subgroup of pulmonary ade­nocarcinomas (WHO 1982) that apparently arise from alveoli or bronchioles. Acinar adenocarcino­mas are another subgroup of pulmonary adeno­carcinomas and may be of bronchial origin (WHO 1982; Dermer 1982). Almost all rat tumors appear to be of the bronchi­olar/alveolar type, although one rat was reported to have a mucinous bronchiolar carcinoma with tall columnar cells and mucus production more analogous to the human acinar adenocarcinoma (Yang and Grice 1965). Bronchiolar/alveolar car­cinomas have a low tendency to metastasize; of 21 pulmonary carcinomas found in 3099 male and female F344 rats, only one metastasized to the me-

diastinum (Goodman et al. 1980). Two other cases of pulmonary tumor metastases have been report­ed in the F344 rat (Reznik 1981). Spontaneous bronchiolar/alveolar tumors are infrequent in most rat strains (Burek and Hollander 1977; Table 7). They are reported to be induced by ex­posure to nickel carbonyl, beryllium sulfate, and chrysotile asbestos (Kuschner and Laskin 1970).

Comparison with Other Species

In man, bronchiolar/alveolar carcinoma is one of the least common of the primary lung cancers, ac­counting for only 5% ofthetotal cases (Greenberg 1982). While the cell of origin has been controver­sial, it appears that both Clara cells and type II pneumocytes can give rise to this tumor (Green­berg 1982; Dermer 1982; Lieber et al. 1976; Singh et al. 1981; Schraufnagel et al. 1982). In the mouse bronchiolar/alveolar carcinomas are quite com­mon, depending upon the strain, and arise from both Clara cells and type II pneumocytes (Kauff­man 1981). In both the dog and cat approximately three-fourths of the pulmonary tumors are classi­fied as adenocarcinomas, while only 26 of 171 pulmonary tumors in dogs and five of 47 pulmo­nary tumors in cats were considered to be of alve­ogenic origin (Moulton et al. 1981). Ultrastructur­al studies suggest that at least some of the rat tumors are of type II pneumocyte origin (Reznik­Schuller and Reznik 1982).

References

Anver MR, Cohen Bl, Lattuada CP, Foster SI (1982) Age­associated lesions in barrier-reared male Sprague-Daw­ley rats: a comparison between Hap: (SD) and Crl: COPS(R)CD(R)(SD) stocks. Exp Aging Res 8: 3-24

Boorman GA, Hollander CF (1973) Spontaneous lesions in the female W AG/Rij (Wistar) rat. 1 Gerontol 28: 152-159

Burek ID, Hollander CF (1977) Incidence patterns of spontaneous tumors in BN/Bi rats. INCI 58: 99-105

Coleman GL, Barthold SW, Osbaldiston GW, Foster SF, lonas AM (1977) Pathological changes during aging in barrier-reared Fischer 344 male rats. 1 Gerontol 32: 258-278

Dermer GB (1982) Origin of bronchioloalveolar carcino­ma and peripheral bronchial adenocarcinoma. Cancer 49:881-887

Goodman DG, Ward 1M, Squire RA, Paxton MB, Rei­chardt WD, Chu KC, Linhart MS (1980) Neoplastic and nonneoplastic lesions in aging Osborne-Mendel rats. Toxicol Appl Pharmacol55: 433-447

Greenberg SD (1982) Histology and ultrastructure ofbron­chiolo-alveolar carcinoma. In: Shimosato Y, Melamed

Page 125: Respiratory System

Bronchiolar/Alveolar Carcinoma, Lung, Rat 115

Table 7. Naturally occurring bronchiolar/alveolar tumors in rats

Strain Sex No. Age Tumors' Reference

Adenomas Carcinomas

ACIIN M 55 Life span 0 0 Maekawa and Odashima 1975

ACIIN F 209 Life span 0 0 Maekawa and Odashima 1975

F344 M 2305 24mo 35 (1.5) 18 (0.8) Haseman et al. 1984 F344 F 2354 24mo 20 (0.9) 9 (0.4) Haseman et al. 1984 F344 M 96 Life span 0 0 Jacobs and Huseby 1967 F344 F 102 Life span 0 0 Jacobs and Huseby 1967 F344 M 160 Life span 0 0 Sass et al. 1975 F344 F 192 Life span 0 0 Sass et al. 1975 F344 M 144 Life span 0 0 Coleman et al. 1977 Holtzman-SD M/F 268 24mo 0 0 MacKenzie and Gamer 1973 Oregon M/F 673 24mo 0 1 (0.1) MacKenzie and Gamer 1973 Osbome-Mendel M 50 24mo 0 0 Radomski et al. 1965 Osbome-Mendel F 50 24mo 0 1 (2) Radomski et al. 1965

Osbome-Mendel M 975 24mo 4 (0.4) 5 (0.5) Goodman et al. 1980 Osbome-Mendel F 970 24mo 2 (0.2) 3 (OJ) Goodman et al. 1980 Sherman M 60 24mo 0 0 Kociba et al. 1974 Sherman F 60 24mo 0 0 Kociba et al. 1974 Sprague-Dawley M 179 18mo 1 (0.6) 2 (1.1) Prejean et al. 1973

Sprague-Dawley F 181 18mo 0 1 (0.5) Prejean et al. 1973

Sprague-Dawley M 85 24mo 1 (1.2) 1 (1.2) Kociba et al. 1978 Sprague-Dawley F 86 24mo 0 0 Kociba et al. 1978

Sprague-Dawley HAP M 45 18-38mo 1 (2.2) 1 (2.2) Anver et al. 1982 (SD)

Sprague-Dawley Crl: M 84 18-38 mo 0 0 Anver et al. 1982 COPS (SD)

Sprague-Dawley M 655 Life span 2 (0.3)b 2 (O.W Ross and Bras 1965 Wistar M 150 24mo 0 0 Torkelson et al. 1974 Wistar F 139 24mo 0 0 Torkelson et al. 1974

Wistar M 472 30mo 0 1 (0.2) Kroes et al. 1981

Wistar F 457 30mo 0 0 Kroes et al. 1981 Wistar F 290 Life span 0 0 Boorman and Hollander

1973

• Total tumors found (%) b All tumors found in the 210 animals receiving high-protein diet

Page 126: Respiratory System

116 Gary A. Boorman

MR, Nettesheim P (eds) Morphogenesis oflung cancer, voI1.CRC, Boca Raton, pp 121-145

Haseman JK. Huff JE, Boorman GA (1984) Use of his tori­cal control data in carcinogenicity studies in rodents. Toxicol Pathol in press

Jacobs BB, Huseby RA (1967) Neoplasms occurring in aged Fischer rats with special reference to testicular, uterine and thyroid tumors. JNCI 39: 303-309

Kauffman SL (1981) Histogenesis of the papillary Clara cell adenoma. Am J Pathol 103: 174-180

Kociba RJ, McCollister SB, Park C, Torkelson TR, Geh­ring PJ (1974) 1,4-Dioxane. I. Results of a 2-year inges­tion study in rats. Toxicol Appl Pharmacol 30: 275-286

Kociba RJ, Keyes DG, Beyer JE, Carreon RM, Wade CE, Dittenber DA, Kalnins RP, Frauson LE, Park CN, Bar­nard SD, Hummel RA, Humiston CG (1978) Results of a two-year chronic toxicity and oncogenicity study of 2,3,7,8-tetrachlorodibenzo-p-dioxin in rats. Toxicol Appl Pharmacol 46: 279-303

Kroes R, Garbis-Berkvens JM, de Vries T, van Nesselrooy HJ (1981) Histopathological profile of a Wistar rat stock including a survey of the literature. J Gerontol 36: 259-279

Kuschner M, Laskin S (1970) Pulmonary epithelial tumors and tumor-like proliferations in the rat. In: Nettesheim P, Hanna MG Jr, Deatherage JW (eds) Morphology of experimental respiratory carcinogenesis, AEC Sympo­sium Series 21 USAEC, Oak Ridge, pp 202-226

Lieber M, Smith B, Szakal A, Nelson-Rees W, Todaro G (1976) A continuous tumor-cell line from a human lung carcinoma with properties of type II alveolar epithelial cells. Int J Cancer 17: 62-70

Mac Kenzie WF, Gamer FM (1973) Comparison of neo­plasms in six sources of rats. JNCI 50: 1243-1257

Maekawa A, Odashima S (1975) Spontaneous tumors in ACI/N rats. JNCI 55: 1437-1445

Moulton JE, von Tscharner C, Schneider R (1981) Classifi­cation oflung carcinomas in the dog and cat. Vet Pathol 18: 513-528

Prejean JD, Peckham JC, Casey AE, Griswold DP, Weis­burger EK. Weisburger JH (1973) Spontaneous tumors in Sprague-Dawley rats and Swiss mice. Cancer Res 33: 2768-2773

Radomski JL, Deichmann WB, MacDonald WE, Glass EM (1965) Synergism among oral carcinogens. Toxicol AppIPharmacoI7:652-656

Reznik G (1981) Unusual sites of lung tumor metastases in B6C3F1 mice and F344 rats. Anticancer Res 1: 159-162

Reznik-SchUller HM, Reznik G (1982) Morphology of spontaneous and induced tumors in the bronchiolo­alveolar region of F344 rats. Anticancer Res 2: 53-57

Reznik-SchUller HM, Hague BR Jr, Creasia DA (1981) Ul­trastructure of pulmonary tumors induced in rats by N­nitrosomethylurethane. J Toxicol Environ Health 8: 501-506

Ross MH, Bras G (1965) Tumor incidence patterns and nu­trition in the rat. J Nutr 87: 245-260

Sass B, Rabstein LS, Madison R, Nims RM, Peters RL, Kelloff GJ (1975) Incidence of spontaneous neoplasms in F344 rats throughout the natural life span. JNCI 54: 1449-1456

Schraufnagel D, Peloquin A, Pare JA, Wang NS (1982) Differentiating bronchioloalveolar carcinoma from ad­enocarcinoma. Am Rev Respir Dis 125: 74-79

Singh G, Katyal SL, Torikata C (1981) Carcinoma of type II pIieumocytes. Am J Pathol102: 195-208

Torkelson TR, Leong BKJ, Kociba RJ, Richter WA, Geh­ring PJ (1974) 1,4-Dioxane. II. Results of a 2-year inhala­tion study in rats. Toxicol Appl Pharmacol 30: 287-298

WHO (1982) Histological typing of lung tumors. Neo­plasma 29: 111-123

Yang YH, Grice HC (1965) Mucinous bronchiolar carci­noma of the rat lung: a case report. Can J Comp Med 29: 15-17

Page 127: Respiratory System

Squamous Cell Carcinoma, Lung, Syrian Hamster 117

Squamous Cell Carcinoma, Lung, Syrian Hamster

Parviz M. Pour and Hildegard M. Reznik-Schuller

Synonyms. Squamous cell tumor; epidermoid car­cmoma.

Gross Appearance

Tumors of 0.5 mm in diameter can be recognized by careful examination of the cut section of the lungs. They may be situated along the bronchi or be randomly distributed all over the lung paren­chyma. There is a tendency of tumors to flow to­gether and in some cases a whole lobe may be in­volved. Small nodules are usually well demarcat­ed, fleshy, and firm and the appearance of their cut surface depends on tumor type. Pure squa­mous cell tumors are often grayish white and granular on the cut surface. Larger tumors may be cystic or may undergo central necroses. In mixed squamous-adenocarcinomas a viscous fluid may be wiped from the cut surface.

Microscopic Features

Histologic demonstration of the lesion can be fa­cilitated by placing individually dissected lobes of the hamster lung in fixative. For histologic pro­cessing the fixed lung should be cut along the course of the main bronchi in order to include the bronchi in the sections. As in the upper respiratory tract, malignant squa­mous cells arise from bronchial or bronchiolar ep­ithelial cells which have undergone metaplasia. These malignant cells, in the form of small or large islands, expand into the surrounding alveoli (Fig. 154), gradually replacing the alveolar epithe­lium and filling out the alveolar lumen. Small foci of tumor cells can often be found within the pul­monary parenchyma, remote from the bronchi (Fig. 155), a finding which may indicate their pri­mary development from alveolar epithelium. However, the possibility that they represent intra­luminal growth from a terminal bronchus or plug­ging of a bronchus by metastases of laryngeal or tracheal lesions cannot always be excluded. Virus­and benzo(a)pyrene-induced tumors have been thought to originate from bronchioli, and others, produced by external radiation, from the alveolar­bronchiolar epithelium. Pulmonary squamous cell carcinomas occur in re­markably varied patterns and include well or

moderately differentiated, keratinizing types (Fig. 156); nonkeratinizing, transitional cell types; mixed glandular, adenosquamous cell or mucoep­idermoid cell types; and anaplastic types. Some tumors present keratin-filled, occasionally large cysts (distended bronchi ?), from the walls of which tumor cells begin to invade (Fig. 157). Squamous cell differentiation can also be ob­served within or in the vicinity of adenocarcino­mas and anaplastic carcinomas (Fig.158), find­ings which support the concept that all these tumors originate from common stem cells that dif­ferentiate into several cell types, including neuro­endocrine cells.

Ultrastructure

Multiple focal hyperplasias in segmental bronchi and bronchioles (Fig. 159) have been identified by electron microscopy as proliferating endocrine (APUD-type) cells [for N-nitrosodiethylamine (DEN) and N-nitrosodibuthylamine (DBN) at weeks 2-3, for N-nitrosomorpholine (NM) at weeks 4-5] (see Table 1 for abbreviations). The characteristic dense-cored granules of these en­docrine cells decreased in number under continu­ed nitrosamine treatment and were replaced by bundles of cytoplasmic tonofilaments (Fig. 160). The formation of these bundles is the ultrastruc­tural marker for early squamous metaplasia. Lung tumors contained areas with ultrastructural fea­tures of endocrine and squamous cells.

Differential Diagnosis

Primary squamous cell carcinomas can be distin­guished from secondary tumors in most but not all cases. The presence of tumor cells in blood ves­sels and subpleural locations favors their metas­tatic nature, but small intraalveolar lesions are at times extremely difficult to distinguish from aspi­rated pharyngolaryngotracheal neoplasms. Aspi­rated tumor cells may evoke inflammatory reac­tions; however, this can also occasionally be seen around primary tumors (Figs. 155 and 160). A de­cision can often be reached by determining the general pattern of induced tumors. Granulomas associated with calcification of the

Page 128: Respiratory System

118 Parviz M. Pour and Hildegard M. Reznik-Schuller

Fig. 154 (Above). Lung, hamster. Squamous metaplasia of a terminal bronchus. Note part of the original bronchus {ar­row}. metaplastic epithelium of the bronchus, and squa­mous cell nests beneath it. Hand E, x 195

Fig. 155 (Below). Lung, hamster. Islands of malignant squa­mous cells within the pulmonary parenchyma with no ob­vious relation to the bronchus {right}. Note inflammatory reaction around the tumor. Hand E, x 78

Page 129: Respiratory System

Fig. 156 (Above). Lung, hamster. Squamous cell carcinoma with limited tendency to keratinize. Hand E, x 195

Squamous Cell Carcinoma, Lung, Syrian Hamster 119

Fig. 157 (Below). Lung, hamster. Beginning of alveolar in­vasion by a well-differentiated and cystic squamous cell carcinoma. Hand E, x 195

Page 130: Respiratory System

120 Parviz M. Pour and Hildegard M. Reznik-Schuller

Fig. 158. Anaplastic carcinoma, lung, hamster, composed oflarge and bizarre cells, one with squamous cell differentiation (arrow). Hand E, x 390

bronchial and bronchiolar basal membranes may simulate pleomorphic tumors (Pour and Birt 1979).

Biologic Features

Squamous cell carcinomas are unknown as a spontaneous entity in Syrian hamsters. Therefore, all our comments refer to tumors experimentally induced by one or more carcinogens.

Pathogenesis. The pathogenesis of bronchial tu­mors induced in Syrian golden hamsters by sub­cutaneous treatment with DEN (see Table 1) (giv­en at the rate of 17.8 mg/kg body wt. twice weekly), DBN (351 mg/kg given once weekly), and NM (98 mg/kg once a week) have been stud­ied in serial sacrifice experiments by light and electron microscopy (Reznik-Schuller 1976, 1977a, b, 1983a, b; Reznik-Schuller and Reznik 1979). Each of these three nitrosamines induced the same spectrum of lesions. However, the laten­cy period was different for each compound. Ultrastructural findings parallel observations of

multidirectional differentiation in human lung tu­mors, in which admixtures of squamous, endo­crine, and glandular features may be found (Churg et al. 1980; Gould et al. 1981). Moreover, the results demonstrate that these nitrosamines can selectively induce multiple proliferations of the otherwise sparse endocrine cells in the ham­ster lung. This model system has been successfully used for the in vitro cultivation of pulmonary en­docrine cells (Linnoila 1982; Linnoila et al. 1981), and makes studies of the cells of origin of human oat cell carcinoma (pulmonary endocrine cells) possible.

Incidence. As with other respiratory tract tumors, the incidence of pulmonary squamous cell carci­nomas varies in relation to the specificity and po­tency of the carcinogen for the pulmonary tissue. Among carcinogens tested N-nitrosodi-n-propyl­amine (DPN) was more effective than its assumed beta-metabolites for inducing this tumor type (Althoff et al. 1977 a; Pour et al. 1973, 1974 a). The most potent carcinogen was N-nitrosovinyl­ethyl amine, (VEN) which induced incidences of 75% squamous cell carcinomas and 17% adeno-

Page 131: Respiratory System

Fig. 159. Segmental bronchus of a hamster after 4 weeks of administration of N -nitrosodiethyl­amine. In the basal epithelial layer, note proliferated cells with ultrastructural features of neuroendocrine (APUD-type) cells. Uranyl acetate and lead citrate. TEM, x 1700

Fig. 160. Cell of segmental bronchus of a ham­ster that received N-nitrosomorpholine for 12 weeks. In this neuroendocrine cell, many of the dense-cored granules have been replaced by tonofilament bundles. Uranyl acetate and lead citrate, TEM, x 12000 (reduced by 15%)

Squamous Cell Carcinoma, Lung, Syrian Hamster 121

Page 132: Respiratory System

122 Parviz M. Pour and Hildegard M. Reznik-SchUller

squamous cell carcinomas (Althoff et al. 1977b). DEN also resulted in induction of a number of pulmonary squamous cell carcinomas (Donte­will and Mohr 1961; Herrold and Dunham 1963; Mohr 1970; Althoff et al. 1971) Other carcino­gens, such as N-nitrosobis(2-acetoxypropyl)amine (BAP) (Pour et al. 1976) and N-nitrosohexame­thyleneimine (N-6-MI) (Althoff et al. 1973), are relatively weak inducers of these neoplasms and N-nitrosomethyl(2-oxopropyl)amine (MOP) (Pour et al. 1980), N-nitrosobis(2-hydroxypro­pyl)amine (BHP) (Pour et al. 1975), N-nitroso-2-oxopropyl-n-propylamine (2-0PPN) (Pour et al. 1974a), N-nitroso-2-hydroxypropyl-n-propyl­amine (2-HPPN) (Pour et al. 1974b), and N-ni­trosomethyl-n-propylamine (MPN) (Pour et al. 1974c) were ineffective, although these carcino­gens, especially BHP, have induced a high inci­dence of papillomas and carcinomas in the upper respiratory tract. Alkylnitrosamines with one me­thyl group in one aliphatic chain in an alphaposi­tion apparently lose their specificity for the lungs, but gain potency for the nasal epithelium (Pour et al. 1980) Among other carcinogens, external ra­diation (DeVilliers and Gross 1966), 9,10-di­methyl-1,2-benzanthracene (DMBA) Gross et al. 1965), and benzo(a)pyrene (BaP) have been re­ported as potent inducers of pulmonary squa­mous cell carcinomas (Herrold and Dunham 1963; Dontenwill and Mohr 1962; Saffiotti 1970; Saffiotti et al. 1964, 1966, 1972a, b; Henry and Kaufman 1973; Henry et al. 1973, 1975; Schreiber et al. 1974; Reznik-Schuller and Mohr 1974, 1975). Viral infections have been reported to be etiologic agents also (Rabson et al. 1960).

Sex. No clear-cut sex differences in terms of tu­mor incidence or predominant tumor types have been observed, either in our series or in those from other laboratories. Squamous cell carcinomas grow expansively. It is difficult to define invasion in tissues such as lungs, which are composed of intercommunicat­ing sacs without limiting capsules. However, in general these lesions, in contrast to adenomas, are not well circumscribed. We consider any squa­mous cell tumor of the lungs to be a carcinoma, regardless of size. Despite their local invasive be­havior, the metastasis of such tumors must be ex­tremely rare, and was not observed in our materi­al.

References

Althoff J, Cardesa A, Pour P, Mohr U (1973) Carcinogenic effect of n-nitrosohexamethylenimine in Syrian golden hamsters. JNCI 50: 323-329

Althoff J, Grandjean C, Pour P, Bertram B (1977 a) Com­parison of the effect of beta-oxidized dipropylnitrosa­mine metabolites administered at equimolar doses to Syrian hamsters. Z Krebsforsch 90: 141-148

Althoff J, Grandjean C, Russell L, Pour P (1977b) Vinyl­ethylnitrosamine: a potent respiratory carcinogen in Syrian hamsters. JNCI 58: 439-442

Althoff J, Wilson R, Mohr U (1971) Diethylnitrosamine­induced alterations in the tracheobronchial system of Syrian golden hamster. JNCI 46: 1067-1071

Churg A, Johnston WH, Stulbarg M (1980) Small cell squamous and mixed small cell squamous-small cell an­aplastic carcinomas of the lung. Am J Surg Pathol 4: 255-263

De Villiers AJ, Gross P (1966) Morphologic changes in­duced in the lungs of hamsters and rats by external radi­ation (x-rays). A study in experimental carcinogenesis. Cancer 19: 1399-1410

Dontenwill W, Mohr U (1961) Carcinome des Respira­tionstraktus nach Behandlung von Goldhamstern mit Diathylnitrosamin. Z Krebsforsch 64: 305-312

Dontenwill W, Mohr U (1962) Vergleichende Untersu­chungen an metaplastichen und malignen Epithelveran­derungen des Respirationstraktes im Tierexperiment. Z Krebsforsch 65: 168-170

Gould VE, Memoli VA, Dardi LE (1981) Multidifferentia­tion in human epithelial cancers. J Submicro Cytol 13: 97-103

Gross P, Tolker E, Babyak MA, Kaschak M (1965) Experi­mental lung cancer in hamsters. Arch Environ Health 11:59-65

Henry MC, Kaufman D (1973) Clearance of benzo(a)py­rene from hamster lungs after administration on coated particles. JNCI 51: 1961-1964

Henry MC, Port CD, Bates RR, Kaufman DG (1973) Re­spiratory tract tumors in hamsters induced by ben­zo( aJpyrene. Cancer Res 33: 1585-1592

Henry MC, Port CD, Kaufman DG (1975) Importance of physical properties of benzo(aJpyrene-ferric oxide mix­tures in lung tumor induction. Cancer Res 35: 207-217

Herrold KM, Dunham U (1963) Induction of tumors in the Syrian hamster with diethylnitrosamine (N-nitroso­diethylamine). Cancer Res 23: 773-777

Linnoila RI (1982) Effects of diethylnitrosamine on lung neuroendocrine cells. Exp Lung Res 3: 225-236

Linnoila RI, Nettesheim P, DiAugustine RP (1981) Lung endocrine-like cells in hamsters treated with liiethylni­trosamine: alterations in vivo and in cell culture. Proc Nat! Acad Sci USA 78: 5170-5174

Mohr U (1970) Effects of diethylnitrosamine in the respi­ratory system of Syrian golden hamsters In: Nettesheim P, Hanna MG, Deatherage JW (eds) Morphology of experimental respiratory carcinogenesis, AEC Sympo­sium Series No.21. USAEC, Div Tech Info Ext, Oak Ridge, pp 255-265

Pour P, Birt D (1979) Spontaneous diseases of Syrian gold­en hamsters - their implications in toxicological re-

Page 133: Respiratory System

search: facts, thoughts and suggestions. Prog Exp Tu­mor Res 24: 145-156

Pour P, KrUger FW, Cardesa A, Althoff J, Mohr U (1973) Carcinogenic effect of di-n-propylnitrosamine in Syrian golden hamsters. JNCI 51: 1019-1027

Pour P, Althoff J, Cardesa A, KrUger FW, Mohr U (1974 a) Effect of beta-oxidized nitrosamines on Syrian golden hamsters. II. 2-0xopropyl-n-propylnitrosamine. JNCI 52: 1869-1874

Pour P, KrUger FW, Althoff J, Cardesa A, Mohr U (1974b) Effect of beta-oxidized nitrosamines on Syrian golden hamster. I. 2-Hydroxypropyl-n-propylnitrosamine. JNCI 52: 1245-1249

Pour P, KrUger FW, Cardesa A, Althoff J, Mohr U (1974 c) Tumorigenicity of methyl-n-propylnitrosamine in Syrian golden hamsters. JNCI 52: 457-462

Pour P, KrUger FW, Althoff J, Cardesa A, Mohr U (1975) Effect of beta-oxidized nitrosamines on Syrian ham­sters. III. 2,2'-Dihydroxy-di-n-propylnitrosamine. JNCI 54:141-146

Pour P, Althoff J, Gingell ~ Kupper ~ KrUger F, Mohr U (1976) n-Nitroso-bis(2-acetoxypropyl)amine as a further pancreatic carcinogen in Syrian golden hamsters. Can­cer Res 36: 2877-2884

Pour P, Gingell ~ Langenbach ~ Nagel D, Grandjean C, Lawson T, Salmasi S (1980) Carcinogenicity of N-nitro­somethyl(2-oxopropyl)amine in Syrian hamsters. Can­cer Res 40: 3585-3590

Rabson AS, Branigan WJ, Legallais FY (1960) Lung tu­mors produced by intratracheal inoculation of polyoma virus in Syrian hamsters. JNCI 25: 937-965

Reznik-Schuller H (1976) Proliferation of endocrine (APUD-type) cells during early N-diethylnitrosamine­induced lung carcinogenesis in hamsters. Cancer Lett 1 : 255-258

Reznik-Schuller H (1977 a) Ultrastructural alterations of APUD cells during early N-diethylnitrosamine-induced carcinogenesis. J Pathol 121: 79-82

Reznik-Schuller H (1977 b) Sequential morphologic altera­tions in the bronchial epithelium of Syrian golden ham­ster during N-nitrosomorpholine-induced pulmonary tumorigenesis. Am J Pathol 89: 59-66

Reznik-SchUller HM (1983 a) Cancer induced in the respi­ratory tract of rodents by N-nitroso compounds. In: Reznik-SchUller HM (ed) Comparative respiratory tract carcinogenesis, vol 2, Experimental respiratory tract car­cinogenesis. CRC, Boca Raton, chap 5

Squamous Cell Carcinoma, Lung, Syrian Hamster 123

Reznik-Schuller HM (1983b) Carcinogens, the pulmonary endocrine cell, and lung cancer. In: Becker K, Gazdar A (eds) Proceedings, The endocrine lung in health and dis­ease. Saunders, Philadelphia

Reznik-SchUller H, Mohr U (1974) Investigations on the carcinogenic burden by air pollution in man. X. Mor­phological changes of the tracheal epithelium in Syrian golden hamsters during the first 20 weeks of benzo( ajpy­rene instillation: an ultrastructural study. Zentralbl Bak­teriol (Orig B) 159: 503-525

Reznik-Schuller H, Mohr U (1975) Investigations on the carcinogenic burden by air pollution in man. XII. Early pathological alterations of the bronchial epithelium in Syrian golden hamsters after intratracheal instillation of benzo(a)pyrene. Zentralbl Bakteriol (Orig B) 160: 108-129

Reznik-Schuller H, Reznik G (1979) Experimental pul­monary carcinogenesis. Int Rev Exp Pathol 20: 211-281

Saffiotti U (1970) Experimental respiratory tract carcino­genesis and its relation to inhalation exposures. In: Hanna MG, Nettesheim P, Gilbert JR (eds) Inhalation carcinogenesis, AEC Symposium Series No 18 (CONF-691001). USAEC Div Tech Info Ext, Oak Ridge TN, pp 27-54

Saffiotti U, Cefis F, Kolb LH (1964) Bronchogenic carci­noma induction by particulate carcinogens. Proc Am Assoc Cancer Res 5: 55

Saffiotti U, Cefis F, Shubik P (1966) Histopathology and histogenesis of lung cancer induced in hamsters by car­cinogens carried by dust particles. In: Severi L (ed) Lung tumours in animals. Div Cancer Research, University of Perugia, Perugia, pp 537-546

Saffiotti U, Montesano ~ Sellakumar AR, Kaufman DG (1972a) Respiratory tract carcinogenesis induced in hamsters by different dose levels ofbenzo(ajpyrene and ferric oxide. JNCI 49: 1199-1204

Saffiotti U, Montesano ~ Sellakumar A~ Cefis F, Kauf­man DG (1972b) Respiratory tract carcinogenesis in hamsters induced by different numbers of administra­tions ofbenzo( ajpyrene and ferric oxide. Cancer Res 32: 1073-1081

Schreiber H, Saccomanno G, Martin DH, Brennan L (1974) Sequential cytological changes during devel­opment of respiratory tract tumors induced in hamsters by benzo(ajpyrene-ferric oxide. Cancer Res 34: 689-698

Page 134: Respiratory System

124 Gary A. Boorman

Squamous Cell Carcinoma, Lung, Rat

Gary A. Boorman

Synonym. Epidermoid carcinoma.

Gross Appearance

The gross appearance of spontaneous squamous cell carcinomas of the rat lung has apparently not been described in the literature. Squamous cell carcinomas induced in the rat lung by polycyclic hydrocarbons are seen grossly as solid masses containing central areas of friable material (kera­tin) (Shabad and Pylev 1970). Naturally occurring squamous cell carcinoma in man tends to arise in the hilar region and is thus more centrally located than other tumor types, which may arise in the pe­riphery of the lung (WHO 1982). Induced squa­mous cell carcinomas of the rat lung often arise at the site of application of the carcinogen. A site predilection for the naturally occurring squamous cell carcinoma of the rat lung has not been report­ed.

Microscopic Features

The spontaneous squamous cell carcinomas found in the lung of the NCIINTP control F344 rats are composed almost entirely of squamous epithelial cells (Fig. 161). The tumor cells often show little atypia (Fig. 162), produce keratin, and show orderly differentiation (Fig. 163). Their ma­lignant nature is evident by invasive growth into the surrounding lung parenchyma and one of the three naturally occurring tumors had metasta­sized to the mediastinum (Fig. 164). Induced squamous cell carcinomas of the rat lung appear more anaplastic. Microscopically the tumors con­tain both well-differentiated squamous epitheli­um and areas of atypical polymorphic cells with hyperchromatic nuclei and numerous mitotic fig­ures (Shabad and Pylev 1970). Both naturally oc­curring and induced squamous cell carcinomas of the rat lung frequently have a marked scirrhous response (Kuschner and Laskin 1970; Shabad and Pylev 1970).

Ultrastructure

A description of the ultrastructural features of the spontaneous squamous cell carcinoma of the rat lung was not found in a review of the literature. In squamous cell carcinomas induced in the lung of the rat by N-nitrosoheptamethyleneimine, ultra­structural features include the formation of tono­filaments, keratohyalin, and keratin with invasive growth into the adjacent lung parenchyma (Rez­nik-Schuller and Gregg 1981).

Differential Diagnosis

Squamous cell carcinoma must be differentiated from squamous cell hyperplasia and also from bronchial alveolar carcinoma containing areas of squamous metaplasia. Rats with vitamin A-defi­cient diet and/or chronic murine mycoplasmosis may have extensive areas of squamous metaplasia and hyperplasia which could be confusing (Kuschner and Laskin 1970). Areas of squamous metaplasia have an orderly progression of differ­entiation from a layer of basal cells undergoing keratinization, have less atypia, and do not dis­rupt or invade normal pulmonary structures as will squamous cell carcinomas. The marked scir­rhous response found in many squamous cell car­cinomas is a useful feature in distinguishing the tumor from metaplasia. Distant metastases or in­vasion confirm the malignant nature of the lesion. Bronchiolar/alveolar carcinomas usually have a glandular pattern and are clearly different. In man, less well-differentiated adenocarcinomas

Fig.161 (Upper left). Squamous cell carcinoma, rat lung. ~ Note abundant keratin production. Hand E, x 80

Fig.162 (Upper right). Squamous cell carcinoma, rat lung. Note little cellular atypia and minimal keratin production. Hand E, x 160

Fig.163 (Lower left). Squamous cell carcinoma, rat lung. Orderly progression of differentiation is evident from basal layer to central area of keratin. Hand E, x 300

Fig. 164 (Lower right). Metastasis of pulmonary squamous cell carcinoma to mediastinum. Cells are less differentiat­ed and mitotic figures are frequent. Hand E, x 350

Page 135: Respiratory System

Squamous Cell Carcinoma, Lung, Rat 125

Page 136: Respiratory System

126 Gary A. Boorman

can be distinguished from squamous cell carcino­mas by different immunohistochemical patterns of keratin staining (Said et al. 1983). In the rat, well-differentiated bronchiolar/alveolar carcino­mas may show areas of squamous cell differentia­tion. Since bronchiolar/alveolar carcinomas rise from different cell types in the periphery of the lung, it is crucial to separate them from the squamous cell carcinomas for proper interpretation of long-term toxicologic studies. Squamous cell carcinomas may be more central in location and should be composed entirely of squamous cells. Tumors showing a glandular or alveolar pattern mixed with squamous areas would be more properly di­agnosed as bronchiolar/alveolar carcinoma with squamous differentiation.

Biologic Features

Induced squamous cell carcinomas of the rat ap­pear to begin with basal cell hyperplasia in the air­ways. Bundles of tonofilaments are later found in the cells in the hyperplastic areas, suggesting squamous cell metaplasia. The squamous cell car­cinomas later develop in the areas of metaplasia (Reznik-Schuller and Gregg 1981). Several studies on radiation-induced squamous cell carcinoma in the rat lung also showed progressive stages from squamous metaplasia to carcinoma (Kuschner and Laskin 1970). Squamous metaplasia of the re­spiratory epithelium precedes or may be a "pre­condition" for squamous cell carcinoma in man (Grundmann 1983; Saccomanno et al. 1970). Since this tumor type is one of the most frequent in man, several models have been developed to produce squamous cell carcinomas in the rat. Placement of a thread laden with methylcholan­threnes in the lung, intratracheal instillation of polycyclic hydrocarbons with absorbents such as India ink powder and carbon black, and implan-

tation of intrabronchial pellets containing poly­cyclic hydrocarbons all produce squamous cell carcinomas in the rat (Deutsch-Wenzel et al. 1983; Kuschner and Laskin 1970; Shabad and Pylev 1970). The induced tumors in the rat simulate broncho­genic carcinoma in man, arising from bronchial epithelium, being locally invasive, and having a tendency to metastasize to hilar lymph nodes and kidneys (Kuschner and Laskin 1970). In contrast to induced mouse pulmonary tumors, which are reported to be weakly immunogenic (Yuhas et al. 1975; Pasternak et al. 1966), squamous cell carci­nomas induced in the rat respiratory tract are ca­pable of eliciting both cellular and humoral re­sponse in immunized isogenic recipients (Jamasbi et al. 1978). In a review of the literature, most arti­cles on naturally occurring tumors of various rat strains reported no squamous cell carcinomas of the lung. A few large series did contain one or more squamous cell carcinomas, as shown in Table 8. In 2-year studies squamous cell carcino­mas of the lung were not found in female rats and occurred at a very low incidence in males (0.1 %-0.2%). In rats allowed to complete their life span a slightly higher incidence was found in males (0.6%) and one squamous cell carcinoma was found in a female rat (Table 8). There is no evidence in this series of any strain differences in incidence - apparently, they are very uncommon in most rat strains.

Comparison with Other Species

Squamous cell carcinoma is a very uncommon type of pulmonary neoplasm in the rat, as op­posed to man, in whom this is the most frequent type of tumor (WHO 1982; Rothschild et al. 1982). In the dog and cat squamous cell carcino­mas account for 6% and 12% of the lung tumors respectively (Moulton et al. 1981). The induced

Table S.N aturally occurring squamous cell carcinoma in the rat lung

Strain Age" Incidenceb

Males Females

Wi star 124 weeks 1/472 (0.2) 0/457 Osbome-Mendel 126 weeks 1/975 (0.1) 0/970 F344 116 weeks 312305 (0.1) 012356 F344 Life span 3/529 (0.6) 1/529 (0.2)

" All reports include animals dying spontaneously: age given is the age at time of death b Incidence equals no of animals with squamous cell carcinomal animals at risk (%)

Reference

Kroes et al. 1981 Goodman et al. 1980 Haseman et. al. 1984 Solleveld et al. 1984

Page 137: Respiratory System

Radiation-Induced Squamous Cell Carcinoma, Lung of Rodents 127

tumors in the rat are reported to have biologic characteristics analogous to those in man (Kusch­ner and Laskin 1970), but spontaneous tumors are rare and their biolgic characteristics have not been established.

References

Deutsch-Wenzel RP, Brune H, Grimmer G, Dettbarn G, Misfeld J (1983) Experimental studies in rat lungs on the carcinogenicity and dose-response relationship of eight frequently occurring environmental polycyclic aromatic hydrocarbons. JNCI 71: 539-544

Goodman DG, Ward JM, Squire RA, Paxton MB, Rei­chardt WD, Chu KC, Linhart MS (1980) Neoplastic and nonneoplastic lesions in aging Osborne-Mendel rats. Toxicol Appl Pharmacol 55: 433-447

Grundmann E (1983) Classification and clinical conse­quences of precancerous lesions in the digestive and re­spiratory tracts. Acta Pathol Jpn 33: 195-217

Haseman JK, Huff JE, Boorman GA (1984) Use of his tori­cal control data in carcinogenicity studies in rodents. Toxicol Pathol in press

Jamasbi RJ, Nettesheim P, Kennel SJ (1978) Demonstra­tion of cellular and humoral immunity to transplantable carcinomas derived from the respiratory tract of rats. Cancer Res 38: 261-267

Kroes R, Garbis-Berkvens JM, de Vries T, van Nesselrooy HJ (1981) Histopathological profile of a Wistar rat stock including a survey of the literature. J Gerontol 36: 259-279

Kuschner M, Laskin S (1970) Pulmonary epithelial tumors and tumor-like proliferations in the rat. In: Nettesheim P, Hanna MG, Deatherage JW (eds) Morphology of experimental respiratory carcinogenesis, AEC Sympo­sium Series 21. USAEC, Oak Ridge, pp 203-226

Moulton JE, von Tscharner C, Schneider R (1981) Classifi­cation of lung carcinomas in the dog and cat. Vet Pathol 18:513-528

Pasternak G, Hoffmann F, Graffi A (1966) Growth of di­ethylnitrosoamine-induced lung tumours in syngeneic mice specifically pretreated with x-ray killed tumour tis­sue. Folia Bioi (Praha) 12: 299-304

Reznik-Schuller HM, Gregg M (1981) Pathogenesis of lung tumors induced by n-nitrosoheptamethyleneimine in F344 rats. Virchows Arch [Pathol Anat] 393: 333-343

Rothschild H, Buechner H, Welsh R, Vial LJ, Weinberg R (1982) Histologic typing of lung cancer in Louisiana. Cancer 49: 1874-1877

Saccomanno G, Saunders RP, Archer VE, Auerbach 0, Brennan L (1970) Metaplasia to neoplasia. In: Nettes­heim P, Hanna MG, Deatherage JW (eds) Morphol­ogy of experimental respiratory carcinogenesis, AEC Symposium Series 21. USAEC, Oak Ridge, pp 63-82

Said JW, Nash G, Banks-Schlegel S, Sassoon AF, Muraka­mi S, Shintaku IP (1983) Keratin in human lung tumors: Patterns of localization of different-molecular-weight keratin proteins. Am J Pathol 113: 27 -32

Shabad LM, Pylev LN (1970) Morphological lesions in rat lungs induced by polycyclic hydrocarbons. In: Nettes­heim P, Hanna MG, Deatherage JW (eds) Morphology of experimental respiratory carcinogenesis, AEC Sym­posium Series 21. USAEC, Oak Ridge, pp 227 -242

Solleveld HA, Haseman JK, McConnell EE (1984) Natu­ral history of body weight gain, survival and neoplasia in the F 344 rat. JNCI 72: 929-940

WHO (1982) Histological typing of lung tumors. Neo­plasma 29: 111-123

Yuhas JM, Pazmiiio NH, Wagner E (1975) Development of concomitant immunity in mice bearing the weakly immunogenic line 1 lung carcinoma. Cancer Res 35: 237-241

Radiation-Induced Squamous Cell Carcinoma, Lung of Rodents

Fletcher F. Hahn

Synonym. Epidermoid carcinoma.

Gross Appearance

Squamous cell carcinomas usually appear as spherical or multilobulated nodules with circum­scribed borders (Fig.165). They may be solitary or, on occasion, several may be found in one lung. Solitary nodules may achieve a relatively large size and occupy much of the thorax. There is no predilection for anyone lung lobe. They are

found in the parenchyma and are rarely oriented around major airways. The nodules are frequently seen as gray-white through the pleura. The pleura may be elevated by the nodule in the deflated lung, but invasion of the pleura is rare. On the cut surface, the nodules are dry, gray-white, and generally firm. In well­differentiated tumors, the cut surface may be dry and caseous.

Page 138: Respiratory System

128 Fletcher F. Hahn

Fig. 165. Squamous cell carcinoma in the rat, note spheri­cal nature and circumscribed borders typical of these tu­mors. F344 rat 588 days after inhalation exposure to an al­pha-emitter, mixed transuranic oxides

Microscopic Features

The well-differentiated forms of squamous cell carcinoma are easily recognized by their mimick­ing of squamous epithelium and the keratiniza­tion of the more differentiated layers (Fig. 166). Frequently, the carcinomas are multilobulated, consisting of nests of readily identifiable squa­mous epithelium, keratohyalin, and epithelial pearls. The epithelium is dysplastic and contains foci of anaplastic cells. These tumors can grow to occupy nearly the entire lung. In some cases, much of the tumor mass comprises necrotic keratinized cells sloughed from a thin border of keratinized squamous epithelium at the periphery of the mass (Fig.168). These tumors are usually solitary with a circumscribed border. Clas­sification of some of these well-differentiated tu­mors as malignant is equivocal, but the masses do grow slowly, compressing surrounding parenchy­ma and compromising pulmonary function. None of the well-differentiated tumors metastasizes or invades the pleura. The poorly differentiated forms of squamous cell carcinoma are still readily recognizable by their

individual cell keratinization (Fig. 167). Small nests of epithelial cells are formed, but there is no semblance of normal maturation of the epitheli­um. At the periphery of the nests, there is usually a layer of large anaplastic cells that may have a small amount of eosinophilic cytoplasm. Toward the center of the nest, the cells usually have more eosinophilic cytoplasm, indicative of individual cell keratinization (Fig. 169). Sometimes there is a fibrous stroma, but the tumors are usually not sclerosing. These tumors grow extensively within the lung and are locally invasive, but rarely metas­tasize or invade the pleura. Metastasis, when it oc­curs, is usually confined to the thorax and local lymph nodes. The invasiveness of these tumors is illustrated by their growth into pulmonary veins, which has been noted in both rats and mice. The invasive tissue is usually poorly differentiated (Fig. 170) but can be well differentiated.

Ultrastructure

The ultrastructure of these tumors has not been studied in rodents. However, ultrastructural char­acterization is not required for diagnosis because of the typical squamous characteristics of even the poorly differentiated tumors.

Differential Diagnosis

Grossly, squamous cell carcinomas must be dif­ferentiated from other lung tumors and from ab­scesses. The poorly differentiated tumors may ap­pear grossly like adenocarcinomas of the lung. The squamous cell carcinomas are generally nod­ular, whereas the adenocarcinomas tend to fill the whole lobe of the lung. The well-differentiated squamous cell carcinomas may appear grossly like benign tumors or abscesses of the lung. Gen­erally, the cut surface of the squamous cell carci­noma has a drier center with a lamellated gray ap­pearance. Abscesses generally have a soft center with homogeneous appearance. Microscopically, squamous cell carcinomas must be differentiated from squamous metaplasia, squamous papillomas, and adenocarcinomas. Squamous metaplasia can be quite extensive in

Fig. 166 (Upper left). Well-differentiated squamous cell car- t> cinoma in the lung of an F 344 rat 588 days after inhalation exposure to a beta-emitter, 144Ce02. Two small foci of keratin formation are present. Hand E, x 100

Page 139: Respiratory System

Radiation-Induced Squamous Cell Carcinoma, Lung of Rodents 129

Fig.167 (Lower left). Poorly differentiated squamous cell carcinoma in the lung of a Syrian hamster 398 days after inhalation of 144Ce02. Many individual cells have abun­dant cytoplasm with keratin. Hand E, x 100

Fig.168 (Upper right). Well-differentiated squamous cell carcinoma in the lung of an F 344 rat 453 days after inhala­tion exposure to mixed transuranic oxides. The periphery of the tumor is well differentiated and has produced

.. . .

~;' :. ,.: ... "',. ". t.,

; " ':': ;~, ,:, • 'J'" ,\:

~ •. :.'" !' I ... : .. , ... • '. • ~ .' \ !.I~

• . :".!' "',

: I

abundant keratin that is accumulated in the center. The tu­mor has compressed the surrounding normal lung paren­chyma. Hand E, x 63

Fig. 169 (Lower right). Poorly differentiated squamous cell carcinoma in the lung of an F 344 rat 557 days after inhala­tion exposure to mixed transuranic oxides. Small nests are present, with a distinct peripheral layer of cells and a cen­tral accumulation of large cells with abundant eosinophilic cytoplasm. Hand E, x 100

Page 140: Respiratory System

130 Fletcher F. Hahn

Fig. 170. Growth of a poorly differentiated squamous cell carcinoma into a pulmonary vein of a C 57 BL/6J mouse 295 days after inhalation exposure to 144Ce02. Such inva­sion of vessels may be seen, but metastasis of radiation-in­duced squamous cell carcinomas outside the thoracic cavi­ty in rodents is rare. Hand E, x 100

the lung, particularly in mice. Metaplasia is usual­ly associated with bronchioles and alveolar ducts, but may extend to peripheral alveoli. Occasional­ly, metaplasia occurs solely in the alveoli, usually in association with focal scars or particulates in the alveoli. Metaplasia is well differentiated, espe­cially in rats, and usually conforms to the normal alveolar architecture of the lung. Some well-dif­ferentiated squamous tumors in rats are large no­dules with a well-differentiated wall of squamous epithelium and a center filled with keratin debris. These have been classified as endophytic papillo­mas. Histologically, they frequently resemble epidermal cysts with little papillary differentia­tion. They may occur as solitary nodules, but fre­quently malignant foci or masses are associated with the papillomas, indicating that malignant change occurs from the papillomas. Poorly differ­entiated squamous cell carcinomas may resemble adenocarcinomas, but individual squamous cells generally have a more abundant, pale eosinophil­ic cytoplasm with an angular outline.

Biologic Features

Rodents have been used extensively in studies of radiation carcinogenesis of the lung using inter­nally deposited alpha- or beta-emitters or external irradiation. Modes of exposure have been inhala­tion, intratracheal instillation, or pellet implanta­tion of radionuclides, or external thoracic irradia­tion. These different modes may affect the type of neoplasia or incidence of neoplasia. A good re­view of radiation carcinogenesis in the respiratory tract has been published by Kennedy and Little (1978). Table 9 gives results reported from studies with ra­dioactive materials in rats. Squamous cell carcino­mas are the most common radiation-induced tu­mors in rats exposed to beta or gamma radiation. Adenocarcinomas, however, occur more fre­quently in rats that inhale alpha-emitting radio­nuclides. This higher incidence may relate to dif­ferences in radiation dose distribution in the lung or to differences in the route of administration, i. e., inhalation vs implantation or instillation. Ra­diation quality factors may also playa role. Masse (1980) has commented on the histogenesis of lung tumors induced in rats by inhalation of al­pha-emitters. He does not state specifically from which studies the examined rats came, but a pre­vious report from the same laboratory indicates that the histologic type of lung tumor was inde­pendent of the radionuclide inhaled: 244Cm, 241Am, 235Pu, 238Pu, or 239Pu (LaFuma et al. 1974). Masse reviewed 500 epithelial lung tumors and classified them according to the World Health Or­ganization classification of human lung tumors. The five most common types were: epidermoid carcinoma (40%), bronchogenic adenocarcinoma (13%), bronchoalveolar carcinoma (36%), com­bined epidermoid and bronchogenic adenocar­cinoma (5%), and large cell carcinoma (1.6%). Transmission electron microscopy was used to ex­amine 34 of the tumors, but improved the diagno­sis only in the case of large cell carcinomas. Sanders and Dagle (1974) noted a direct correla­tion between the degree of inflammation and sclerosis around a radioactive particle and squa­mous metaplasia in surrounding tissue, ftequently in subpleural regions. They envisioned progres­sion from squamous metaplasia to squamous cell carcinoma. Masse (1980), however, did not find scarring and metaplasia to be a prerequisite for the development of such carcinomas. Table 10 was prepared from studies with radioac­tive materials in mice. Squamous cell carcinomas in mice are induced infrequently by irradiation.

Page 141: Respiratory System

Radiation-Induced Squamous Cell Carcinoma, Lung of Rodents 131

Table 9. Lung tumor types in rats exposed to radioactive materials'

Compound Exposure Percent prevalenceb Range of doses to lung (rad) Squamous Adeno- Adenoma Other

Other

Internal emitters Alpha-emitters

Particulates 238Pu 239Pu 244cd.. or 253Es

Gases RnandRn daughters

Beta-emitters Implants 32p 90Sr 106Ru ' Particulates 35S, 144Ce Particulates 144Ce

External irradiation

Inhalation

Inhalation

Implant

Intratracheal instillation Inhalation

7 - 10 x 1Q3

38 - 92 x 10Z WLMc

0.34- 16 x 1<r

24 -200 x 104

0.3 - 74 x 1<r

cell carcinoma carcinomas

<1-9 7-36

56 3

17-58

13-23

14-36 1-8

<1-3

2

Hemangio­sarcoma Fibrosarcoma Mesothelioma

Lymphosarcoma

Reticulosarcoma

X-rays Thoracic 0.3 - 0.5 x 1<r 10 43 5 Fibrosarcoma

• Compiled from Annals of ICRP (1980) and Gracey et al. (1979); ranges are the range of averages for all the studies on any particular group of compounds

b Percent prevalence = percentage of animals at risk that develop tumor type noted C WLM, working level month; although estimates vary, 1 WLM a.; 0.5-1 rad

Table 10. Lung tumor types in mice exposed to radioactive materials'

Compound Exposure Percent prevalenceb Range of doses to lung (rad) Squamous Adeno- Adenoma Other

Internal emitters Alpha-emitters

239PU02

Beta-emitters Particulates 106Ru02 Particulates 9OY,144Ce

Gamma-emitters 6OCo wire

External irradiation X-rays

Intratracheal instillation

Intratracheal

Inhalation

Implant

Thoracic

120- 4000

300- 9000

990-32000

9- 46x1<r

750- 2750

cell carcinoma carcinomas

5 400 Fibrosarcoma

4C 75c Lymphosarcoma

2 <1-3c 9-13c Hemangio-sarcoma Fibrosarcoma

10

23c

• Compiled from Annals of ICRP (1980), Hahn et al. (1980) and Lundgren et al. (1981); ranges are the range of averages for all the studies on any given group of compounds

b Percent prevalence = percentage of animals at risk that develop tumor type noted c Increase over control

Page 142: Respiratory System

132 Fletcher F. Hahn

Table 11. Lung tumor types in Syrian hamsters exposed to radioactive materials"

Compound

Alpha-emitters Particulates 21OpO, Fe203 Particulates 238Pu, 239Pu Gases Rn and Rn daughters

Beta-emitters Particulates 144Ce02

Exposure mode

Intratracheal instillation Inhalation

Inhalation

Inhalation

Range of doses to lung (rad)

300-5000

66-9000

85- 120 x 102 WLMC

60- 500 x 102

Percent prevalenceb

Squamous Adeno- Adeno- Adenoma cell carcinoma squamous carcinomas carcinoma

3 97

2 <1-14 <1-21

2

3 2

a Compiled from Annals of ICRP (1980), Cross et al. (1981), Lundgren et al. (1982), Lundgren et al. (1983) and Thomas et al. (1981) - ranges are the range of averages for all the studies on any given group of compounds

h Percent prevalence = percentage of animals at risk that develop tumor type noted C WLM, working level month; although estimates vary, 1 WLM ~ 0.5-1 rad

Table 12. Comparison of squamous cell carcinoma characteristics in laboratory ro­dents and humans exposed to radiation

Characteristic Rats Mice Syrian Mana hamsters

Prevalence <1%-60% 0-10% 2%-3% 3% Differentiation Good Moderate Poor Poor Metastasis Rare Rare Rare Frequent Site of origin Small Small Small Large

airways, airways, airways, airways alveoli alveoli alveoli

a Based on data from uranium miners (Horacek et al. 1977 and Kunz et al. 1979)

The occurrence of these tumors in any study seems related to high radiation dose to the lung, for example with the 60Co wire implants. Table 11 contains results reported from studies with radioactive materials in Syrian hamsters. It is difficult to induce lung tumors in Syrian hamsters with radiation. The lone exception is the intratra­cheal instillation of 210pO and hematite. In this model, 15 weekly intratracheal instillations are given. Tumors can be induced in as little as 15 weeks, and by 1 year essentially all hamsters have tumors.

Comparison with Other Species

Squamous cell carcinomas of the lung of different species of laboratory rodents are histologically similar. There are differences, however, in several characteristics, as noted in Table 12. More differ-

ences are apparent between squamous cell carci­nomas in laboratory rodents and those in man. Rarely are the tumors in laboratory rodents as poorly differentiated or aggressive as in man, where distant metastases are common. In man, squamous cell carcinomas most frequently origi­nate in major airways; this is rare in rodents. This difference may be related to differences in the site at which the radiation dose is delivered.

Acknowledgements. Research was performed un­der U. S. Department of Energy Contract Number DE-AC04-76EV01013. Research was conducted using facilities fully accredited by the American Association for the Accreditation of Laboratory Animal Care.

Page 143: Respiratory System

References

Annals of the ICRP (1980) Biological effects of inhaled ra­dionuclides. Pergamon, New York

Cross FT, Palmer RF, Busch RH, Filipy RE, Stuart BO (1981) Development of lesions in Syrian golden ham­sters following exposure to radon daughters and urani­um ore dust. Healthy Phys 41: 135-153

Gracey DR, Fish JE, Divertie MB (1979) Experimental squamous cell lung tumors in Sprague-Dawley and mu­rine pneumonitis-free rats. Cancer 44: 558-603

Hahn FF, Lundgren DL, McClellan RO (1980) Repeated inhalation exposure of mice to 144Ce02. II. Biologic ef­fects. Radiat Res 82: 123-137

HoracekJ, Placek V, SevcJ (1977) Histologic types of bronchogenic cancer in relation to different conditions of radiation exposure. Cancer 40: 832-835

Kennedy AR, Little JB (1978) Radiation carcinogenesis in the respiratory tract. In: Harris CC (ed) Pathogenesis and therapy of lung cancer. Dekker, New York

Kunz E, SevcJ, Placek V, Horacek J (1979) Lung cancerin men in relation to different time distribution of radiation exposure. Health Phys 36: 699-706

La Fuma J, Nenot JC, Morin M, Masse R, Metivier H, No­libe D, Skupinski W (1974) Respiratory carcinogenesis in rats after inhalation of radioactive aerosols of acti­nides and lanthanides in various physiochemical forms.

Pleural Mesothelioma, Syrian Hamster 133

In: Karbe E, ParkJF (eds) Experimental lung cancer: carcinogenesis and bioassays. Springer, Berlin Heidel­berg New York

Lundgren DL, Hahn FF, McClellan RO (1981) Toxicity of 90y in relatively insoluble fused aluminosilicate particles when inhaled by mice. Radiat Res 88: 510-523

Lundgren DL, Hahn FF, McClellan RO (1982) Effects of single and repeated inhalation exposure of Syrian ham­ster to aerosols of 144Ce02. Radiat Res 90: 374-394

Lundgren DL, Hahn FF, Rebar AH, McClellan RO (1983) Effects of the single or repeated inhalation exposure of Syrian hamsters to aerosols of 239PU02. Int J Radiat Bioi 43: 1-18

Masse R (1980) Histogenesis of lung tumors induced in rats by inhalation of alpha emitters: an overview. In: Sanders CL, Cross FT, Dagle GE, Mahaffey JA (eds) Pulmonary toxicology of respirable particles. DOE sym­posium series, 53 CONF-791 002. National Technical In­formation Service, Springfield

Sanders CL, Dagle GE (1974) Studies of pulmonary car­cinogenesis in rodents following inhalation of transu­ranic compounds. In: Karbe E, ParkJF (eds) Experi­mental lung cancer: carcinogenesis and bioassays. Springer, Berlin Heidelberg New York

Thomas RG, Drake GA, London JE, Anderson EC, PrineJR, SmithDM (1981) Pulmonary tumours in Syrian hamsters following inhalation of 239Pu02. Int J Radiat Bioi 40: 605-611

Pleural Mesothelioma, Syrian Hamster

Antonio Cardesa and Josep A. Bombi

Synonyms: Malignant mesothelioma; mesothelial neoplasia.

Gross Appearance

Pleural mesotheliomas appear as whitish gray no­dules, usually multiple and of firm consistency. When initially detected they form tiny masses of less than 1 mm in diameter, which progressively grow and fuse together to form larger nodules and plaques measuring up to 5 mm at the widest point. The development of these nodules takes place from the parietal as well as from the visceral pleu­ra, extending along and covering the pleural sur­faces. The pleural cavity may be filled by the growth of the mesothelioma; nevertheless, even in advanced lesions, gross invasion of the lung par­enchyma is as a rule not evident. In some meso­theliomas the pleural thickening may be particu­larly striking at the interlobular fissures. These

lesions, when seen from the cut surface of the lung, may give a false impression of tumor inva­sion. Pleural effusions are occasionally present.

Microscopic Features

Three main histological types of mesotheliomas are recognized: epithelioid, fusocellular, and mixed. The epithelioid mesotheliomas are made up of large polyhedral cells, with abundant am­phophilic cytoplasm and sharply defined cell con­tours. Their nuclei are vesiculated, rounded to oval in shape, with the chromatin mainly distrib­uted adjacent to the nuclear membrane or at­tached to it. Frequently, epithelioid mesothelio­mas give rise to the development of papillary formations (Figs. 171 and 172), which are support­ed by thin stalks of connective tissue. In other cases, epithelioid mesotheliomas are manifested initially in nodular forms (Fig. 173). Occasionally,

Page 144: Respiratory System

134 Antonio Cardesa and losep A. Bombi

Page 145: Respiratory System

<l Fig.171 (Upper left). Papillary mesothelioma, Syrian ham­ster, at early stage of development, growing at the pleural surface. Hand E, x 100

Fig. 172 (Upper right). Papillary mesothelioma, Syrian ham­ster. Epithelioid cells with mitotic figures. Hand E, x 400

Fig.173 (Lower left). Mesothelioma, hamster, attached to the pleural surface. Hand E, x 100

Fig.174 (Lower right). Epithelioid mesothelioma, Syrian hamster, with spreading pattern. Hand E, x 100

Fig.175 (Above). Epithelioid mesothelioma, Syrian ham- C>

ster. Cells with eccentric nuclei and a lymphoplasmacytoid appearance. Hand E, x 400

Fig.176 (Lower left). Fusocellular mesothelioma, Syrian hamster. Note cells with spindle-shaped cytoplasm imitat­ing the pattern of fibrosarcoma. Hand E, x 200

Fig.177 (Lower right). Fusocellular mesothelioma, Syrian hamster. Note spindle-shaped cells with nuclei elongated in the direction of the cellular axis. Hand E, x 400

Pleural Mesothelioma, Syrian Hamster 135

Page 146: Respiratory System

136 Antonio Cardesa and Josep A. Bombi

the epithelioid type of mesothelioma may have a tubular or gland-like arrangement of complex spaces, which mimic the features of adenocarci­nomas. In most instances, it is possible to see how these tumor cells originate from the mesothelial covering (Fig. 174); in other cases they seem to de­velop from the submesothelial layer. Epithelioid mesotheliomas may grow in the form of wide sheets. In many areas these are composed of com­pactly arranged cells, in others the cells may be loosely attached to one another or even be devoid of intercellular cohesiveness. The nuclei of these cells may be eccentrically located, sometimes con­ferring upon them a plasmacytoid resemblance (Fig. 175). The fusocellular type of mesothelioma is com­posed of cells with spindle-shaped cytoplasm, poorly defined cell contours, and oval nuclei elongated in the direction of the main cellular axis (Figs. 176 and 177). Fusocellular mesotheliomas are able to form collagen, imitating the histologi­cal pattern of fibrosarcomas. This resemblance is occasionally striking, since they may even adopt a herringbone structural arrangement. The cells fre-

Fig. 178. Epithelioid mesothelioma, Syrian hamster. Tumor cells within lymphatic vessels. Hand E, x 200

quently have atypical mitoses. Fusocellular meso­theliomas may also be arranged in intertwining or tangled strands, thus mimicking malignant fi­brous histiocytomas. Mesotheliomas of the mixed pattern have areas with characteristic epithelioid features alternating with other areas in which the fusocellular pattern is quite evident. In the hamster, the type of mesothelioma most commonly observed is epithelioid. These epithe­lioid cells are PAS positive, except after diastase digestion, and positive as well for alcian blue, but the color fades after treatment with hyaluroni­dase. The mesothelioma cells do not seem to con­tain mucin.

Ultrastructure

The ultrastructural features of mesotheliomas have been reported in other laboratory rodents (Davis 1979; Kawai 1979) but we are not aware of reports on the ultrastructural features of pleural mesotheliomas in the hamster.

Differential Diagnosis

The main differential diagnoses include neopla­sias metastatic to the pleura, particularly carcino­ma and sarcoma. In these instances, the correct diagnosis should be based on finding a primary neoplasm with the histologic features of the tu­mor found in the pleura. In distinguishing epithel­ioid mesotheliomas from metastatic carcinomas, the presence or absence of mucin in the pleural tumors is helpful because this substance is not produced by mesotheliomas. Another diagnosis to be considered is malignant lymphoma extend­ing through the mediastinum, lung, and pleura. The epithelioid cells of mesothelioma, particu­larly when they lack cohesiveness, may adopt plasmacytoid features that may mimic a plasma­cytoma or lymphoplasmacytic lymphoma. The absence of lymphoma in other organs should argue against that diagnosis. Finally, malignant mesotheliomas should not be confused w;ith reac­tive fibrous pleural plaques, which are fibroblastic proliferations producing abundant collagen, usually in the vicinity of asbestos fibers.

Biologic Features

Natural History. Most mesotheliomas manifest their malignant behavior by superficial growth

Page 147: Respiratory System

along the pleura. Invasion of the underlying chest wall or lung parenchyma is usually minimal or discrete in comparison to the tendency for intra­cavitary growth. A mesothelioma tends to fill the pleural cavities, resulting in considerable thicken­ing and even fusion of both pleural surfaces. This may cause compression of lung parenchyma, which is accentuated by the development of pleu­ral effusions. In advanced cases, progressive inva­sion of mediastinum, pericardium, and dia­phragm may occur. Lymphatic vascular invasion (Fig. 178), metastases to regional lymph nodes, or distant metastases should be regarded as excep­tional findings.

Pathogenesis. Mesotheliomas are a well-recog­nized tumor entity and epithelioid mesotheliomas are histologically quite similar to the cells lining the mesothelial surfaces. In spite of this, con­troversy exists with regard to their histogenesis. Some investigators maintain that they arise from cells of the mesothelial lining, others believe that they originate from the underlying submesothelial cells (Alvarez-Fernandez and Diez-Nau 1979; Cardesa et al. 1980). In tHe hamster, due to the paucity of studies, there is not yet enough infor­mation available to add to knowledge on this un­settled issue.

Etiology. Mesotheliomas are produced by expo­sure to different kinds of asbestos and glass fibers.

Frequency, Natural Occurrence, and Experimental Induction. Mesotheliomas in the hamster are in­duced tumors. In a study of spontaneous tumors in two hamster colonies (Pour et al. 1976) no mesotheliomas were observed. Spontaneous mesotheliomas were not recorded in a wide re­view of the literature (Mohr 1982). Rabson et al. (1960) were able to induce mesotheliomas by treatment of hamsters with virus and asbestos. Smith et al. (1964, 1965) induced mesotheliomas by intrapleural injections of amosite into ham­sters. Although Smith et al. (1970) induced pul­monary neoplasias and pleural mesotheliomas by the combined instillation of chrysotile and ben­zopyrene into the lower respiratory tract of ham­sters, no tumors were found when the instillation was of chrysotile alone. More recently, F. Pott (personal communication) has been able to pro­duce pleural mesotheliomas in hamsters by the in­halation of crocidolite and various types of glass fibers.

Pleural Mesothelioma, Syrian Hamster 137

Comparison with Similar Lesions in Human and Other Species

From the morphological standpoint, mesothelio­mas induced in the hamster have striking gross as well as microscopic similarities with their counter­parts in man (Brenner et al. 1982) and in other lab­oratory species (Pott et al. 1976).

References

Alvarez-Fernandez E, Diez-Nau MD (1979) Malignant fibrosarcomatous mesothelioma and benign pleural fi­broma (localized fibrous mesothelioma) in tissue cul­ture: a comparison of the in vitro pattern of growth in re­lation to the cell of origin. Cancer 43: 1658-1663

Brenner J, Sordillo PP, Magill GB, Golbey RB (1982) Ma­lignant mesothelioma of the pleura: review of 123 pat­ients. Cancer 49: 2431-2435

Cardesa A, Alvarez T, Pott F, Huth F, Mohr U Sept (1980) Morphological patterns of mesotheliomas produced by intraperitoneal exposure to various fibres. Abstracts XIllth international congress International Academy of Pathology, p 277

Davis JM (1979) The histopathology and ultrastructure of pleural mesotheliomas produced in the rat by injections of crocidolite asbestos. Br J Exp Pathol 60: 642-652

Kawai TO (1979) Histopathological studies on experimen­tally induced pulmonary, pleural and peritoneal neo­plasms in mice by intraperitoneal injection of chrysotile asbestos and N-methyl- N-nitrosourethane. Acta Pathol Jpn 29: 421-433

Mohr U (1982) Tumours of the respiratory tract. In: Turu­sov VS (ed) Pathology of tumours in laboratory animals, Vol III, Tumours of the hamster. IARC Sci Publ No 34, Lyon, pp115-145

Pott F, Dolgner R, Friedrichs KH, Huth F (1976) L'effect oncongene des poussieres fibreuses. Ann Anat Pathol (Paris) 21: 237-246

Pour P, Mohr U, Cardesa A, Althoff J, Kmoch N (1976) Spontaneous tumors and common diseases in two col­onies of Syrian hamsters. II. Respiratory tract and diges­tive system. JNCI 56: 937-948

Rabson AS, Branigan WJ, Legallais FY (1960) Lung tu­mors produced by intratracheal inoculation of polyoma virus in Syrian hamsters. JNCI 25: 937 -965

Smith WE, Miller L, ChurgJ, SelikoffU (1964) Pleural reaction and mesothelioma in hamsters injected with as­bestos. Proc Am Assoc Cancer Res 5: 9 (abstract #234)

Smith WE, Miller L, Churg J et al. (1965) Mesotheliomas in hamsters following intrapleural injection of asb~stos. J Mt Sinai Hosp (NY) 32: 1-8

Smith WE, Miller L, Churg J (1970) An experimental mod­el for study of carcinogenesis in the respiratory tract. In: NettesheimP, HannaMG, DeatherageJW (eds) Mor­phology of experimental respiratory carcinogensis. AEC symposium series no 21. USAEC, Division of Technical Information Extension, Oak Ridge

Page 148: Respiratory System

138 Bernard Sass and Annabel G. Liebelt

Metastatic Tumors, Lung, Mouse

Bernard Sass and Annabel G. Liebelt

Synonym. Secondary tumors of the lungs.

The spread of cancers to distant organs and the subsequent development of new foci present ma­jor challenges in research on tumor progression. Reports of spontaneously occurring mouse mam­mary tumors metastasizing to the lung first ap­peared in the early years of this century. Experi­ments with induced hepatic tumors and several types of transplanted tumors were reported but the pathogenesis of metastasis was still not under­stood. Metastasis involves several steps: detach­ment of cancer cells, either singly or in clumps, from the primary tumor; invasion of and subse­quent passage within the vascular system to distant sites in which adhesion to and invasion and penetration of the vessel wall occur; and infil-

Fig. 179. Large, multinodular metastasis in lung from spon­taneous mammary tumor in strain RIll female mouse. x 15

tration and growth in the pulmonary tissues. These steps are at present being investigated and will result in a better understanding of the patho­genesis of metastasis.

Gross Appearance

There are few authoritative morphological de­scriptions of metastatic tumors of the lungs of mice (BorreI1903; Murray 1908; Ashburn 1937; Dunn 1945; Brooks 1970). Mouse lungs bearing metastases may have single or multiple nodular growths distributed in one or more lobes, and there is often bilateral involvement. Brooks (1970) described mammary tumor metastases as circum­scribed groups of nodules, surrounded by narrow zones of compressed lung tissue. Tumor nodules may be found deep within the parenchyma of the lung as well as on the pleural surface. Stewart and associates (1970) described the gross appearance of alveologenic carcinomas as sharply circum­scribed pearly white nodules which may project above the pleural surface of the lung. In the experience of the authors, metastatic mam­mary tumors are usually soft and friable, grayish white, and highly vascular, and have a raised nod­ular surface (Fig.179); metastases of mammary tumors with extensive squamous metaplasia or keratinization may appear pearly white. Hepatic tumor metastases are firm, round, pale brown­gray, and well circumscribed (Vesselinovitch et al. 1978). Metastases of primary tumors from con­nective tissues, cartilage, or bone, on the other hand, are usually dense, firm to very hard, rela­tively avascular, white, and shiny. Transplants of malignant melanoma may form metastases in the lungs as either pigmented (black, shiny, and rub­bery) or nonpigmented (white, firm) tumors, de­pending on whether the transplant was pigmented or not (Figs.180 and 181). Although with most lymphoreticular neoplasms the lungs / are in­volved, the lungs often do not contain grossly visi­ble discrete nodules but instead contain a diffuse infiltrate of tumor cells in the entire lung, causing a loss of normal pink color. Heston (1940), using a dissecting microscope, enumerated tumors in the lungs of mice. Several other methods for the detection and enumeration of lung tumors have been described. Wexler

Page 149: Respiratory System

(1966) devised a method for the identification of tumors utilizing the injection of India ink into the trachea of an anesthetized mouse; the ink is al­lowed to flow into the alveolar spaces by gravity. The dissected lungs are washed in tap water and then immersed in Fekete's solution (Fekete 1938). This solution bleaches the neoplastic tissue (Fig. 182), sharply outlining the gross tumors. Subsequently, the lobes of the lung are separated and sliced and the metastatic tumors are identi­fied and counted. Histological identification and enumeration of metastatic tumors are improved by employing serial sectioning of the lung. Ashburn (1937) found 178 of 480 (37%) mammary-tumor-bearing mice to have gross metastases. He cited Murray (1908), who found 39.6% gross metastases in 68 tumor-bearing mice, Haaland (1911), who found 38% in 237, and Marsh (1929), who found 39.1% in 314. However, when Ashburn examined the 178 gross lesions, 31 of these (17.4%) were spurious metas­tases, that is, lung adenomas, lymphoid deposits, abscesses, foci of bronchopneumonia, or other circumscribed inflammatory lesions. Further­more, when examining grossly negative lungs, Ashburn found 70 (14.6%) microscopic metas­tases. Murray (1908) and Marsh (1927) identified, by serial section techniques, metastases in eight of 16 and nine of 13 grossly normal lungs respective­ly. A study by Consolandi and associates (1958) re­vealed macroscopic pulmonary metastases mi­croscopically confirmed in 34% of the cases and, in addition, 46% microscopic metastases in appar­ently normal lungs. The authors did not state which sectioning techniques were used. All of these studies point out the difficulty of relying on­lyon data based on gross observations. Kyriazis et al. (1974) and Vesselinovitch et al. (1978) studied the percentage of pulmonary me­tastases from hepatic tumors induced by diethyl­nitrosamine (DEN). Their technique was to re­move the lungs en bloc with the trachea, thymus, paratracheal lymph nodes, and thyroid glands, and fix these tissues in 10% neutral buffered for­malin. The specimens, including the structures mentioned above, were processed whole, and 5-llm sections were cut at the long axis at two dif­ferent levels, one of which passed through the hi­lar region. Kyriazis et al. (1974) found a metastatic percentage of 21.6% for animals receiving DEN at 1 day of age and 22.9% in animals receiving DEN at 15 days of age. Histologic examination of single, randomly selected lung sections yielded a

Metastatic Tumors, Lung, Mouse 139

metastatic percentage of only 4.3% for males and 0.0% for females. These studies demonstrate the importance of ex­amining adequate sections of all lobes of the lung to obtain the exact number of tumor metastases.

Fig. 180 (Above). Pulmonary metastases (arrows) from sub­cutaneous transplant of melanotic melanoma in (BALBI c x DBAf)Fl hybrid mouse. Note a small amelanotic por­tion of the transplant (arrow). x 2

, Fig.181 (Below). Pulmonary metastases from subcutane­ous transplant of amelanotic melanoma in (BALBI c x DBAf)Fl hybrid mouse. This transplant line was derived by selection of an amelanotic portion from the melanotic transplant line. x 2

Page 150: Respiratory System

140 Bernard Sass and Annabel G.Liebelt

1111111111111111111111111111111 Fig. 182. Lungs of strain RIll mouse with metastatic mam­mary tumor from a single mammary tumor. The lungs were distended with India ink; numerous pulmonary nodules are bleached white following exposure to Fekete's solu­tion. x 2.5

Microscopic Features and Differential Diagnosis

One of the most important biologic criteria in the expression of primary malignant neoplasms is the ability of tumor cells to invade surrounding nor­mal tissues, including the blood and lymph ves­sels. It is this vascular invasion that makes the first stage of metastasis, namely the formation of tu­mor cell emboli, a unique and essential prerequi­site for the spread of malignant tumors (Willis 1973). Mouse tumors metastasizing to the lungs do so mainly by the hematogenous route (Ash­burn 1937; Dunham and Stewart 1953; Consolan­di et al. 1958; Kyriazis et al. 1974; Vesselinovich et al. 1978). There are three main anatomic sites in which tu­mor cells are found in the lungs. The first is within larger blood vessels in the form of tumor cell em­boli or thrombi (Figs.183-185). Although tumor cell thrombi or emboli provide evidence of malig­nancy, they are not considered to be metastases (Willis 1973) until the constituent tumor cells penetrate vessel walls. The second site in the lungs is outside larger vessels but surrounding bronchi (Fig. 183) and within alveoli and alveolar capilla­ries. The third anatomic pattern which is common in metastatic tumors of the lungs of mice is that of disseminated small or large multiple foci in which the relationship to vessels is lost (Fig. 186).

The tumors invade veins rather than arteries, pre­sumably because veins have thinner walls. Most blood-borne metastases are carried by the vena cava to the right heart and thence to the pulmon­ary artery. Tumor cells enclosed by vessel walls are considered to be emboli when they are nonad­herent to vessel walls and thrombi when they are adherent. Tumor cell thrombi may contain other blood ele­ments, such as leukocytes and thrombocytes. Not all tumor emboli or thrombi produce viable me­tastases (Fidler 1980). Those that do so may lodge in capillaries or larger vessels, the walls of which are penetrated by tumor cells. Having gained en­try to pulmonary vessels, there are two main sites at which secondary tumors become established within the lung. The first, intraalveolar, is a frequent site of exten­sion. In mice, alveolar architecture is lost early. Tumor cells invade alveolar spaces and fill and distend alveoli, the walls of which are destroyed by compression. The second site is interstitial ex­tension, and includes invasion of lymphatics, veins, arteries, and bronchi; this is not often evi­dent in mice, except those with primary lung tu­mors that spread within the lung. Tumors which establish in the lung by other than the hematoge­nous route have been reported by several authors (Furth 1946; Epstein 1966). These authors inject­ed leukemia or carcinoma cells intratracheally, bringing about leukemia or carcinoma in the lung. There is a histological similarity between primary alveologenic (alveolar/bronchiolar) tumors and pulmonary metastases from adenocarcinomas of the mammary gland and from tumors of other or-

Fig.183 (Upper left). Lung, mouse, metastases from a sub- t> cutaneous sarcoma, not otherwise specified. Note peri­bronchial distribution of growths, upper left and center. A branch of the pulmonary artery (upper center) contains tu­mor cells. Hand E, x 54

Fig. 184 (Upper right). Lung, mouse, mammary tumor cell emboli in longitudinally sectioned pulmonary artery. The surrounding parenchyma is normal and the integrity of the vessel wall is intact. Hand E, x 80

Fig.185 (Lower left). Orbit, mouse, contents of the orbit with retina below, eyelid above and to left. Harderian gland to right and soft tissues between. The harderian gland is largely replaced by a carcinoma that has invaded a nearby vein (arrow). Hand E, x 54

Fig. 186 (Lower right). Lung, two large metastases of hepa­tocellular carcinoma at either end of the photomicrograph and a small one in the midsection. Note compression of alveoli and invasion of alveolar walls. Hand E, x 38

Page 151: Respiratory System

Metastatic Tumors, Lung, Mouse 141

Page 152: Respiratory System

142 Bernard Sass and Annabel G. Liebelt

gan sites (kidneys, pancreas, uterus) which makes it difficult to identify the primary site (Reznik 1983). Other primary tumors of the mouse lung, such as squamous cell carcinoma, rarely occur spontaneously but have been induced by intra­tracheal instillation of methylcholanthrene. Alveologenic carcinoma develops in the alveolar walls from type II alveolar epithelial cells, which are cuboidal to columnar and are arranged in a glandular pattern often exhibiting papillary for­mations (Stewart et al. 1970, 1979; Stewart 1975). Adenomatosis (see chapter by Boorman, Bronchi­olar/ Alveolar Hyperplasia) is a proliferative and inflammatory lesion which needs to be distin­guished from alveologenic or metastatic tumors. Adenomatosis may be differentiated from alveo­logenic tumors by the following criteria: (a) the pleura is depressed, (b) the alveolar architecture is preserved, and ( c) mucus-containing ciliated or non ciliated columnar and cuboidal cells line the walls of the alveoli (Stewart et al. 1970). Adenom­atosis may follow infection and recovery from Sendai virus. When extrapulmonary primary neoplasms occur singly, their metastases in the lung are readily identified provided that they reproduce the same cellular and architectural patterns as in the pri­mary tumor. When, however, primary tumors of two or more different sites yield single or multiple pulmonary metastases, or if the pulmonary metas­tases differ in morphology from the primary tu­mor, it may be difficult to ascribe the origins of the metastases. In reticulum cell neoplasm type B and lymphocyt­ic leukemia, nodular deposits may be found in the lungs of mice surrounding both vessels and bron­chi. Reticulum cell neoplasm, type B, described by Dunn (1954) and Dunn and Deringer (1968), forms cuffs of tumor cells which surround bron­chi and nearby vessels. Lungs of mice with lym­phocytic leukemia, in addition, often contain in­travascular plugs and deposits of neoplastic lymphocytes in alveolar capillaries. A differential diagnosis must be made distinguishing these pat­terns of hematopoietic tumors from peribronchial and perivascular cuffing due to proliferating mac­rophages and infiltration by lymphocytes follow­ing lesions of chronic pneumonia. Hepatocellular carcinoma and adenocarcinoma of the mammary gland are two tumors that fre­quently occur in inbred mice and commonly me­tastasize to the lung. The morphology of mam­mary gland tumors varies widely within and between strains; indeed, it varies between tumors

within one mouse and within individual tumors. Mammary tumors which metastasize to the lung usually have the same predominant histologic pattern as the parent tumor, and multiple tumors of different histologic types may produce metas­tases that correspond to the primary tumors (Fig. 187). Brooks (1970) reported that a group of metastatic nodules from mammary tumors may become surrounded by a zone of compact tissue containing smaller metastatic tumor nodules mixed with cells of lung origin. Pitelka et al. (1980a, b) examined 160 primary mammary tu­mors by both light and electron microscopy; 69 of these metastasized to the lung and the metastases were confirmed histologically (see also Ultra­structure). In describing the pulmonary metas­tases, the authors stated that "sections of large metastatic tumors often suggest a close resembl­ance to a primary tumor in the same mouse but al­most as frequently do not." Otherwise, the mor­phology of the mammary epithelial cells in the primary and secondary sites was similar. Examination of serial paraffin and epon thick sections of grossly visible pulmonary metastases revealed tumor deposits within the arterioles ac­companying bronchioles (Pitelka et al. 1980b). The tumor invaded the pulmonary connective tis­sue by penetrating the muscular and elastic layers of affected arterioles. Hepatocellular carcinomas induced by carcino­gens are more likely to metastasize than spontane­ous tumors (Figs.188 and 189) (Vesselinovitch et al. 1978). The increased frequency of metastasis of hepatocellular carcinoma is related to trabecular and undifferentiated patterns (see Biology sec­tion). Pulmonary metastases from liver tumors in­duced by DEN (Kyriazis et al. 1974; Vesselino­vitch et al. 1978) were usually multiple and often of microscopic size, and closely resembled those of the primary tumor. There was the suggestion that isolated foci may have coalesced to form larger de­posits. The alveolar capillaries contained tumor cell masses and the resultant growth occasionally distorted bronchiolar lumina (Fig. 188) and com­pressed the surrounding lung tissue (Fig. 189). The pulmonary metastases from hepatocellular carcinomas reduplicate the histologic pattern of the primary tumor. The rare hepatoblastoma can be differentiated from hepatocellular carcinoma by several chracteristics, including: (a) deep stain­ing in hematoxylin and eosin preparations, (b) presence of spindle cells and rosettes, ( c) presence of peculiar "organoids," and (d) presence of hep­atocellular carcinoma in the same section (Turu­sov and Takayama 1979).

Page 153: Respiratory System

Fig.187 (Upper left). Lung, mouse, metastatic mammary adenocarcinoma, type B compresses surrounding alveoli. Hand E, x 54

Fig. 188 (Upper right). Lung, mouse, large metastatic hepa­tocellular carcinoma that has elevated the pleura and com­pressed the lung parenchyma and wall of the alveolar duct. Hand E, x 54

Fig. 189 (Below). Higher magnification of section shown in Fig. 188. Hand E, x 220

Metastatic Tumors, Lung, Mouse 143

Page 154: Respiratory System

144 Bernard Sass and Annabel G. Liebelt

<l Fig.190 (Above). Lung, mouse, metastasis of ovarian gran­ulosa cell tumor that has replaced most of the pulmonary tissue of the apex of the lobe. The pleura is mineralized. H and E, x 54

Fig. 191 (Below). Higher magnification of section shown in Fig. 190. The neoplastic cells are arranged as small nests. H and E, x 130

Fig.192 (Upper left). Lung, mouse, metastasis of renal pel- ~ vic transitional cell carcinoma. The tumor cells are ar­ranged as nests. Hand E, x 220

Fig.193 (Upper right). Lung, mouse, metastasis of renal adenocarcinoma composed of small, closely packed cells with indistinct borders arranged as sheets and nests. Hand E, x 130

Fig.194 (Lower left). Lung, mouse, metastasis of renal ad­enocarcinoma that has compressed and invaded alveoli. Glands are absent. The tumor cells have sharply defined borders, pale-staining cytoplasm, and eccentrically placed, often multiple, nuclei. Hand E, x 220

Fig.195 (Lower right). Lung, mouse, metastasis of harderi­an gland carcinoma. The tumor cells have pale-staining, often vacuolated cytoplasm. Hand E, x 220

Metastases of hepatocellular carcinomas must be differentiated from metastases of adrenal cortical carcinomas and from metastases of granulosa cell tumors. In metastases of adrenal cortical carcino­mas and of granulosa cell tumors (Figs. 190 and 191) the cells tend to be arranged in groups invest­ed by a fine fibrovascular stroma. Granulosa cell tumors with glomerate or tubular pattern may be differentiated from the metastases of adrenal cor­tical carcinoma. Two mice treated with 2-fluorenylacetamide had transitional cell carcinomas of the urinary bladder which metastasized. Both primary tumors had as­sociated squamous cell metaplasia (Frith et al. 1981), but it was not clear whether the metastases had a squamous component. A renal pelvic tran­sitional carcinoma produced metastases com­posed of nests (Fig. 192). Histologically, it may be difficult to dIstinguish metastases of renal adenocarcinomas (Figs.193 and 194) and ofharderian gland tumors (Fig. 195) from primary alveologenic tumors. Pulmonary metastases of renal adenocarcinomas may pro­duce different patterns depending on the patterns of the primary tumor. In the BALB/c/Cf/Cd mouse strain, renal adenocarcinoma cells are small, have scant cytoplasm, and have poorly de-

Page 155: Respiratory System

Metastatic Tumors, Lung, Mouse 145

Page 156: Respiratory System

146 Bernard Sass and Annabel G. Liebelt

Fig. 196 (Upper left). Lung, mouse, metastasis of malignant schwannoma in what remains of vessel wall. Tumor cells have poorly defined borders. Hand E, x 330

Fig.197 (Upper right). Lung, mouse, multiple t~rombi of malignant schwan noma adherent to wall of pulmonary vessel. Hand E, x 330

Fig. 198 (Below). Lung, mouse, pleural metastasis of osteo­sarcoma (A) and adjacent alveologenic carcinoma (B). H and E, x 130

Page 157: Respiratory System

fined borders (Fig. 193). In BALB/c mice the oth­er type of renal adenocarcinoma induced by chemicals (Fig.194) is composed of tumor cells that are larger and have eccentric nuclei, abun­dant, almost clear cytoplasm, and clearly defined cell borders. Spontaneous harderian gland neoplasms in un­treated mice, reported by Sheldon et al. (1983), were classified as adenocarcinoma or one of four different types of adenoma. Of the 30 mice with adenocarcinomas, the tumors of three metasta­sized to the lung and these had a histologic pat­tern similar to that of the primary tumor. The pri­mary tumors of two of these three mice and of two other mice had spread to the periorbital tissues. The authors suggested that periorbital invasion precedes occurrence of distant metastases (Fig. 185). Primary tumors of harderian glands and their metastases (Fig.195) have the distin­guishing feature that the tumor cells may contain intracytoplasmic vacuoles (Sheldon et al. 1983). Pulmonary metastases from sarcomas, especially those that are undifferentiated, form prominent cuffs around blood vessels and bronchi (Fig. 183). With hemangiosarcomas (hemangioendothelio­mas) in the lung it may be difficult to determine whether they are primary or metastatic or repre­sent one site of involement by a tumor of multi­centric origin. Frith et al. (1981) assigned a metas­tatic origin to hemangiosarcomas in the lungs of seven mice treated with 2-fluorenylacetamide, on the basis of finding pulmonary emboli and mil­iary distribution of tumor. Malignant schwannomas, metastatic to the lung, consist almost wholly of Antoni type A tissue, namely interlacing parallel bundles and fibrils (Fig. 196). Lacking were other features of primary schwannoma, including palisading of the nu­clei, Verocay bodies, cysts, and lipoid-contain­ing pseudoxanthomatous cells. The pulmonary vessels contained mUltiple adherent thrombi (Fig. 197). The pulmonary metastases of osteosarcomas ex­hibit all characteristics of the parent tumor, in­cluding bone, osteoid, and spindle cells or undif­ferentiated cells. Among the sections filed in the Registry of Experimental Cancers are those of a mouse with a primary osteosarcoma of bone that had metastasized to the lung, in which there was also a primary alveologenic tumor. The two types of tumor were intermingled (Fig. 198).

Metastatic Tumors, Lung, Mouse 147

Ultrastructure

Examination by electron microscopy of type B mammary tumor metastases in the lung by Brooks (1970) confirmed the acinar pattern of the tumor cells and revealed virus particles budding from cell surfaces, within the gland lumens, and also in some type B (II) cells of the pulmonary alveoli. Wide spaces containing fibrillar material were present at intervals between cells. The mammary tumor cells in the lung were separated from the alveolar airspace by the following: (a) a thick bas­allamina of the mammary tumor cells, (b) a loose connective tissue space containing fibroblasts and collagen fibrils, (c) the basal lamina of the alveo­lar epithelial cells, and (d) the alveolar cells them­selves. Hyperplastic type B (II) pulmonary alveo­lar cells arranged in the form of acini were present in proximity to the metastatic nodules. Cystic spaces lined by mammary tumor cells in some metastatic nodules contained both membranous and fibrillar material. This material was believed by Brooks (1970) to have been synthesized by the tumor cells. Pitelka et al. (1980a, b) compared the ultrastruc­ture of the junctions of the epithelial cells and basal lamina of normal mammary gland tissues, hyperplastic alveolar nodules (HANs), primary mammary carcinomas, and their pulmonary me­tastases. Tight junctions, characteristic of epithe­lial tissues and of normal nonlactating glands, were demonstrated between cells of the neoplastic glands. A feature of the tumors, not present in normal mammary tissue, was the presence of mi­crolumina in otherwise solid deposits. These mi­crolumina were characterized by tight junctions and apical microvilli. Also present in the tumor tissue at varying distances from tight junction belts but not in normal tissue were common mac­ular tight junctions. The authors concluded that since the neoplastic cells have tight junctions, it cannot be proposed that a generalized reduction of adhesive incapability attributable to faculty junctions is a necessary characteristic of malig­nant epithelial cells. Normal basal lamina functions as a barrier be­tween the normal adult epithelium which secretes it and the connective tissue; it also serves as a bar­rier limiting access of macromolecules (Fawcett 1981). Thus it is normally not penetrated except by migrating leukocytes and macrophages. Pitel­ka et al. (1980b) examined the basal lamina ofpri­mary and metastatic mammary tumors, and ob­served that hypertrophy of the basal lamina was the most common structural abnormality of the

Page 158: Respiratory System

148 Bernard Sass and Annabel G. Liebelt

Table 13. Pulmonary metastases from tumors in untreated mice

Organ of origin Diagnosis No. of mice Per- Sex Strain Reference with cent metastasis No. of mice with tumors

Liver Hepatoma 1-2 Slye et al. (1915) Hepatoma 11 17 6 M,F CBA Gorer (1940) Hepatoma 11 97 1 M CF-l Turusov et al.

(1973 b) Hepatoblastoma 01 3 0 M CF-l Turusov et al.

(1973 b) Hepatocellular 43/349 12 M B6C3HF1 Ward et al. (1979) carcinoma 71 58 12 F B6C3HF1 Ward et al. (1979)

51 50 10 M B6C3HF1 National Toxicology Program (1982 a)

01 50 0 F B6C3HF1 National Toxicology Program (1982a)

Mammary Carcinoma nos. 1041273 38 Haaland (1911) gland 261 68 40 Murray (1908)

40 Marsh (1927) 62 (following massage) Marsh (1929)

123/314 39 217/480 45 Infrequent

16 69/160 43

78/464 17 391 80 49 65/169 39 161 78 20 231 58 40 181 61 30

A, B, mixed AB 401 51 78 161 28 57 121 26 46

Carcinoma, nos. 121 19 63

11 6 17 Harderian Adenocarcinoma 31 30 10 gland

nos, not otherwise specified

tumor cells; interruptions were rare, except where necrosis occurred. The hypertrophy was evident as extensive folding, mUltiple layering or irregu­larly increased thickness and could be found: (a) in primary tumors, between stroma and mam­mary tumor epithelial cells, (b) between invasive primary tumors and surrounding connective tis­sue, and (c) in metastases between neoplastic mammary epithelium and tissues of the lung. The authors (Pitelka et al. 1980a) concluded that

Marsh (1929) NIH white Ashburn (1937) AIBrA van der Valk (1981) BALB/cfC3H van der Valk (1981) C3H,C3Hf Pitelka et al. (1980a) BALB/cfC3H BALB/cNIV A., GR, and RIll RIll Liebelt et al. (1968) C3H Liebelt et al. (1968) A Liebelt et al. (1968) DBAl2 Liebelt et al. (1968) RIIIllmr (breeder) Liebelt et al. (1981) RIIIllmr (virgin) Liebelt et al. (1981) BALB/cflC3H/Cb/Se Consolandi et al. RIIIIDmlSe (1958) C3H/Cb/Se Consolandi et al.

(1958) BALB/cfC3H Squartini and

Bistocchi (1977) BALBI cfRIII BALB/C, C3H/He Sheldon et al. (1983) and C57BLl6J

neoplastic mammary epithelium maintains an ef­fective basal lamina barrier while invading non­epithelial tissues. Intravascular metastases con­tained basal lamina interposed between tumor cells and pulmonary vascular endothelium, or if such epithelium ruptured, the basal lamina was found between the tumor cells and the elastica. Following rupture of the arteriolar wall, basal lamina was found between tumor cells and pul­monary connective tissue.

Page 159: Respiratory System

Metastatic Tumors, Lung, Mouse 149

Table 14. Pulmonary metastases from induced tumors in mice

Tissue of Diagnosis No. of mice Per- Treatment ~ex Strain Reference origin with cent

metastasis No. of mice with tumors

Liver Trabecular 221 102 22 DEN at 1 day of age M,F C57BLI Kyriazis et al. (1974) carcinoma 6Jx Trabecular 271 118 23 DEN at 15 days of age M,F C3HeBI Kyriazis et al. (1974) carcinoma FEJ hybrid Trabecular 2661 733 36 One of: BaP, ENU, B6C3F1 Vesselinovitch et al. carcinoma benzidine 2-HCl (1978) Trabecular 52/1076 5 2-FAA in Purina meal F BALBI Frith et al. (1981) carcinoma cStCr!

fC3H/Nctr Hepatoblastoma 0 Controls M,F CF-1 Turusov et al. (1973 a) Hepatoblastoma 4 DDT 2 ppm M,F CF-1 Turusov et al. (1973 a) Hepatoblastoma 0 DDT10ppm M,F CF-1 Turusov et al. (1973 a) Hepatoblastoma 4 DDT 50 ppm M,F CF-1 Turusov et al. (1973 a) Hepatoblastoma 11 DDT 250 ppm M,F CF-1 Turusov et al. (1973 a) Hepatoma 1 Controls M,F CF-1 Turusov et al. (1973 a) Hepatoma 2 DDT 2 ppm M,F CF-l Turusov et al. (1973 a) Hepatoma 2 DDT 10 ppm M,F CF-l Turusov et al. (1973 a) Hepatoma 1 DDT 50 ppm M,F CF-l Turusov et al. (1973 a) Hepatoma 1 DDT 250 ppm M,F CF-1 Turusov et al. (1973 a) Trabecular M,F B6C3F1 National Toxicology carcinoma Program (1982 a) Trabecular 41 49 8 DEHA 12000ppm M B6C3F1 National Toxicology carconoma Program (1982a) Trabecular 51 49 10 DEHA 25000 ppm M B6C3F1 National Toxicology carcinoma Program (1982a) Trabecular 61 49 12 DEHA 12000 ppm F B6C3F1 National Toxicology carcinoma Program (1982a) Trabecular 51 48 10 DEHA 25000 ppm F B6C3F1 National Toxicology carcinoma Program (1982a) Hepatocellular B6C3F1 National Toxicology carcinoma Program (1982b) Hepatocellular 71 49 14 DEHP 12000 ppm M B6C3F1 National Toxicology carcinoma Program (1982b) Hepatocellular 51 50 10 DEHP 25000 ppm M B6C3F1 National Toxicology carcinoma Program (1982b) Hepatocellular 11 50 2 DEHP 12000 ppm F B6C3F1 National Toxicology carcinoma Program (1982b) Hepatocellular 71 50 14 D EHP 25000 ppm F B6C3F1 National Toxicology carcinoma Program (1982b) Trabecular 21 8 25 2,7-FAA 250 ppm for M (DBAl2 Takayama (1968) carcinoma 3-5mo xC57BL)Flo

(DBF1)

Trabecular 31 10 30 F (DBAl2 Takayama (1968) carcinoma xC57BL)Flo

(DBF1)

Trabecular 21 15 14 M (DBAl2 Takayama (1968) carcinoma xC57BL)Flo

(DBF1)

Trabecular 51 16 31 F (DBA12 Takayama (1968) carcinoma xC57BL)Flo

(DBF1)

Trabecular 13 Dieldrin M,F CF-1 Thorpe and Walker carcinoma (1973)

Page 160: Respiratory System

150 Bernard Sass and Annabel G. Liebelt

Table 14 (continued)

Tissue of Diagnosis No. of mice Per- Treatment Sex Strain Reference origin with cent

metastasis No. of mice with tumors

Trabecular 61 Dieldrin M,F CF-1 cited by Vesselinovitch et carcinoma al. (1978) Trabecular 21 9 22 Nafenopen M Acatal- Reddy et al. (1976) carcinoma asemic Trabecular 31 12 25 Nafenopen F CSb Reddy et al. (1976) carcinoma

Mammary Tumors' 491 517 10 2-FAA F BALBI Frith et al. (1981) gland cStCrl

Other tumors fC3H/Nctr Osteogenic 61 13 46 2-FAA F BALBI Frith et al. (1981) sarcomab cStCrl

fC3H/Nctr Renal 41 20 20 2-FAA F BALBI Frith et al. (1981) carcinoma cStCrl

fC3H/Nctr Fibrosar- 31 18 17 2-FAA F BALBI Frith et al. (1981) comab cStCrl

fC3H/Nctr Myoepithe1io- 161 153 11 2-FAA F BALBI Frith et al. (1981) rna cStCrl

fC3H/Nctr Undifferen- 41 72 6 2-FAA F BALBI Frith et al. (1981) tiated cStCrl sarcomab fC3H/Nctr Leiomyosar- 11 36 3 2-FAA F BALBI Frith et al. (1981) comab cStCrl

fC3H/Nctr Granulosa cell 41 197 2 2-FAA F BALBI Frith et al. (1981) tumor cStCrl

fC3H/Nctr Adrenocorti- 31 181 2 2-FAA F BALBI Frith et al. (1981) calcarcinoma cStCrl

fC3H/Nctr Ovary Spleen Angiosarcoma 71 452 2 2-FAA F BALBI Frith et al. (1981) Skin cStCrl

fC3H/Nctr Squamous cell 11 76 1 2-FAA F BALBI Frith et al. (1981) carcinomab cStCrl

fC3H/Nctr Harderian Tumor 2312400 1 2-FAA F BALBI Frith et al. (1981) gland cStCrl

fC3H/Nctr Urinary Transitional cell 21 722 <1 2-FAA F BALBI Frith et al. (1981) bladder carcinoma cStCrl

fC3H/Nctr

DEN, diethylnitrosamine; BaP, benzo (a) pyrene; ENU, ethylnitrosourea; benzidine, p-diaminodiphenyl; '2-FAA, N-2-fluorenylacetamide; 2,7-FAA, N, N' -2, 7 -fluorenylenebisacetamide; DDT, 1,1, 1-trichloro-2,2-bis (p-chlorophenyl)ethane; DEHA, di (2-ethylhexyl)-adipate; DEHP, di (2-ethylhexyl)-phthalate; Dieldrin, 1,2,3,4,10,10-hexachloro-6,7-epoxy-1,4,4 a,5,6,7 ,8,8 a-octohydroendo-exo-1,4 :5,8-dimethanonaphthalene; Najenopen, 2-methyl-2- [P-(1,2,3,4-tetrahydro-1-naphthyl) phenoxy )proprionic acid • 4% adenocarcinoma type A, 73% adenocarcinoma type B, 10% adenocarcinoma type C, 14% adenoacanthoma b Site of primary tumor not given

Page 161: Respiratory System

Biologic Features

Frequency of Metastases

Only a few reports contain data on the frequency with which spontaneous and induced tumors me­tastasize to the lung. Table 13 contains data deal­ing with metastases from spontaneous mammary, liver, and harderian gland tumors. The early reports (Borrell 1903; Bashford and Murray 1904; Murray 1908; Haaland 1911; Marsh 1927, 1929; Pybus and Miller 1934; Ash­burn 1937; Dunn 1945, 1953) on pulmonary me­tastases of spontaneous mammary cancers dealt with percentages and histological appearance. Ashburn (1937) contrasted various percentages of metastasis by whether or not they were detected grossly or histologically in order to eliminate nonmetastatic lesions, while the studies of Conso­landi and associates (1958) established the inci­dence of mammary tumor metastases to the lung for inbred strains of mice with high mammary cancer rates. The percentage of induced hepatic tumors that metastasized to the lungs ranged from 2% to 61 %, with most groups falling between 10% and 20% (Turusov et al. 1973 a; Kyriazis et al. 1974; Vesse­linovitch et al. 1978; Frith et al. 1981). Table 14 lists these and several other reports of pulmonary metastases from induced hepatic tumors. Turusov et al. (1973 a) found a low incidence of metastasis from hepatomas and from hepatoblastomas in mice treated with DDT. Frith and associates (1981) also reported metastatic lesions from 14 types of neoplasms in carcinogen-treated mice (Table 14). A low incidence of pulmonary metas­tasis from mammary and other sites was reported by Turusov et al. (1973 a). Metastasis has been reported for various trans­planted tumors, either as "spontaneous" spread from local transplants (Dunham and Stewart 1953; Stewart et al. 1959; Fisher und Fisher 1967; Liebelt and Liebelt 1967; Liebelt et al. 1968) or as from "experimental" systems, such as intravenous inoculation of tumor cells (Fidler 1975; Miller and Heppner 1979; Fidler and White 1981). Transplantable tumors which produced pulmo­nary metastases included spontaneous or induced tumors in several different inbred strains of mice (Dunham and Stewart 1953). The primary tumors were fibrosarcomas, sarcomas (not otherwise specified), osteogenic sarcomas, rhabdomyosar­comas, melanomas and reticulum cell tumors, and epithelial tumors of the glandular stomach, salivary gland, ovary, testis, cervix, mammary

Metastatic Tumors, Lung, Mouse 151

gland, and skin. The tumors did not often metas­tasize and occurred late in life. Liebelt and Liebelt (1967) developed a model sys­tem for lung metastasis utilizing a transplantable melanoma of (BALBI c x DBAl2f)F1 hybrid mice which, when transplanted subcutaneously, metas­tasized as early as the second transplant genera­tion; successive transplantation of melanotic or amelanotic tumor lines yielded metastases of the same histologic type as the parent tumor. The fre­quency of metastasis of mammary tumors ob­tained from eight mice of two inbred strains and three types of F1 hybrids when transplanted seri­ally by the subcutaneous route for up to 144 gen­erations ranged from 0 to 55% (Liebelt et al. 1968). Under the following headings, we discuss exam­ples of specific host-tumor interrelationships and other factors that have been associated with the occurrence of metastasis.

Tumor Cell Heterogeneity: Tumor cell heteroge­neity is a term used to refer to differences in mor­phology, immunogenicity, growth rate, metabo­lism, hormone receptors, pigment production, ra­diosensitivity, and susceptibility to cytotoxic drugs (Fidler 1978; Fidler et al. 1978), Fidler and Kripke (1977), using clones of the B16 melanoma, showed that cells derived from these clones, when injected intravenously, differed dramatically in their ability to establish tumors in the lung. They obtained similar results when cells from an ultra­violet-induced fibrosarcoma cloned in the same manner were injected intravenously. Grdina et al. (1978), who also used fibrosarcoma cells grown in culture and injected intravenously, demonstrated that the intrinsic clonogenic ability of these cells is independent of cell size and age. More important is the ability of a malignant cell to relocate to a site distant from the primary cancer and there be retained and allowed to proliferate (Grdina et al. 1978). The results of Fogel et al. (1979) indicated that cells of tumors with a high metastatic capaci­ty and distinct antigenic properties exist within the tumor cell popUlation and that immunoselec­tion might be involved in the occurrence of lung metastases. Other investigators (Heppner et al. 1978; Nicolson et al. 1978; Talmadge et al. 1979) have since demonstrated heterogeneity for several rodent tumor cell lines. Thus, it appears that mu­rine neoplasms of both long and short origins are heterogeneous, suggesting the presence of several subpopulations within a given cell line. More recently, Fidler and Kripke (1980) cloned sub lines of uniform highly metastatic cells from

Page 162: Respiratory System

152 Bernard Sass and Annabel G.Liebelt

single lung metastases. This demonstrated that metastases consist of a more homogeneous popu­lation of cells with high metastatic capability than the cells of the primary transplant line.

Genetic Factors (Strain). The frequency of metas­tasis in mammary-tumor-bearing mice of several inbred strains varies depending on the strain (Consolandi et al. 1958; Liebelt et al. 1968).

Histologic Appearance of the Primary Tumor. Ash­burn (1937) divided the primary mammary tu­mors of 185 mice into six histologic groups. The percentages of metastasis to the lung for each his­tologic type were as follows: 51.6% cystadenocar­cinoma, 41.2% adenocarcinoma, 24.2% papillary cystadenocarcinoma, 10.0% "adenoma malig­num," and no cystadenoma, suggesting that the cystadenocarcinomas are clinically more malig­nant than the tumors in the other groups. The studies of Consolandi et al. (1958) disclosed dif­ferent percentages of pulmonary metastases de­pending on the histologic pattern of the primary mammary tumor: type B had 75%, type A 58%, and mixed types A and B had 62%. Apolant (1906), cited by Dunn (1953), pointed out that one could not determine from the histologic examination of a mammary tumor whether it was likely to metastasize to the lungs. More recently, however, van der Valk (1981) reported that 16% of spontaneous mammary tumors of BALB/cfC3H mice metastasize and that such tumors appear less well differentiated than the spontaneous mam­mary tumors in BALBI c mice. In animals surving to the age of 81-90 weeks, 51 % of the hepatocellular carcinomas with a trabecu­lar pattern metastasized to the lung (Vesselino­vitch et al. 1978), irrespective of the chemical car­cinogen that induced them. By contrast, only 1.3% of tumors with an adenomatous pattern metasta­sized, while lesions designated by the authors as hyperplastic nodules of clear, eosinophilic, or ba­sophilic types did not.

Type of Murine Mammary Tumor Virus. The type of murine mammary tumor virus was reported to influence the frequency of metastasis (Squartini and Bistocchi 1977).

Number of Primary Tumors. Metastasis was more frequent in mice having multiple primary mam­mary cancers than in mice with single tumors (Ashburn 1937); for mice with five to eight tu­mors, the rate of metastasis was 70%, for mice with four tumors 58.8%, for mice with two tumors

46.6%, and for mice with a single tumor 37.3%. Consolandi et al. (1958) reported frequency of metastasis to be 83% and 60% respectively in mice with multiple tumors and in mice with a single tu­mor.

Size/Weight of Primary Tumor. Ashburn (1937) re­ported a correlation between the frequency of me­tastasis and the size of the spontaneous primary mammary tumor, expressed as the square root of the product of two dimensions in square millime­ters. Tumors of 10 mm2 or less, according to the above formula, had an incidence of 6.4%, tumors of 10.1-15 mm2 an incidence of 32.5%, and tu­mors of30.1-35 mm2 an incidence of69%. Ander­son et al. (1974) demonstrated that a large in­crease in size and weight of the primary mammary tumor rather than its period of growth correlated with the frequency of metastasis. They found that tumors weighing 0.1-0.9 g metastasized at a rate of 2%, those of 0.9-2.0 g at 23%, and those of 2-4 g at 38%. Large tumors weighing 4.1-17 g me­tastasized at a rate of73%. Several other investiga­tors have reported a direct correlation between the incidence of metastasis and the size of trans­plantable mouse mammary tumors (Coman 1953; Wood et al. 1954; Wexler et al. 1965; Sugarbaker and Cohen 1972).

Duration of Primary Tumor. Although it is difficult to ascertain accurately the date of tumor appear­ance, investigators have established their own cri­teria for establishing the earliest reliable measure­ment. Ashburn (1937) found that the incidence of metastasis increased with duration of tumor; fre­quency of metastasis ranged from 20.7% at 11-20 days to 76.5% when over 100 days.

Surgical Ablation. Liebelt and associates (1968) studied the occurrence of pulmonary metastases of spontaneous mammary tumors in strain A mice 60 days after finding a 1.0 x 1.0 em tumor. The in­cidence in untreated tumor-bearing mice was 39%, in sham-operated mice it was 50%, in mice in which the primary tumor was surgically re­moved it was 80%, and in mice with surgical re­moval and ovariectomy it was 42%.

Location of Primary Tumor. Attempts to correlate anatomic site with frequency of metastasis of mammary tumors gave negative results (Williams et al. 1935; Ashburn 1937; Liebelt et al. 1968). On the other hand, Consolandi et al. (1958) found metastatic frequencies of 71 % and 63% for mam­mary tumors located in the cervical and thoracic

Page 163: Respiratory System

glands respectively, and 62% and 55% for those located in the inguinal and abdominal glands re­spectively.

Growth Rate and Passage. Ashburn (1937) studied the growth rate of single tumors in mice. The me­dian growth rate of tumors that metastasized was well above that of those that had not. There was an exception, however, with tumors that had been present more than 70 days. Growth rate and me­tastatic frequency were correlated in the experi­ments of Consolandi et al. (1958). Heppner and Miller (1981), using transplanted mammary tu­mor cell lines throughout ten transplant genera­tions, were unable to correlate metastasis and bio­logic characteristics such as growth and latent period.

Hormonal Factors. Breeding influenced the fre­quency of metastases of mammary tumors in two strains; that is, breeders had more metastases than virgins (Consolandi et al. 1958; Liebelt et al. 1968; Liebelt et al. 1981) in spite of the fact that virgins -who lived longer developed tumors later than breeders - (Liebelt et al. 1968; Liebelt et al. 1981). Overall breeders developed more tumors (Liebelt et al. 1968). Estrogens and pituitary isografts in­creased the incidence of metastasis. Other hormones, such as growth hormone, adre­nocorticotrophic hormone, and adrenal steroids, promote metastases (Fidler 1975, 1976). Fidler summarized several experiments in which corti­sone promoted the growth of tumor cells in the lungs of mice with transplantable mammary can­cer, other mouse tumor systems, and intravenous­ly injected melanoma cells. One explanation pro­posed is that glucocorticoids may alter the capillary endothelial surface, which may lead to increased stickiness and consequent arrest of tu­mor emboli (Fidler 1976).

Drugs. Lung metastases were enhanced by the cy­tostatic drugs methotrexate, cytoxan, and 5-fluor­ouracil (Heppner et al. 1978).

Cell Surface Properties. It is now accepted that changes in the surface properties of tumor cells are important in determining aspects of growth, invasion, and metastasis (Poste and Weiss 1976). Alterations in the surface properties of the cells of the primary tumor contribute to their escape from many restrictions, for instance growth-regulating factors to which normal cells are subject. These factors could include hormones or serum factors that exert their effects by binding to the cell sur-

Metastatic Tumors, Lung, Mouse 153

face. Recently, differences were found in lectin­binding properties between pairs of high and low metastatic murine tumor lines (Dennis et al. 1981; Kerbel et al. 1982; Altevogt et al. 1983). Altevogt et al. (1983) suggested that one could theoretically envision at least three steps in the metastatic cas­cade that involve cell surface carbohydrate inter­actions: (a) the release of tumor cells from the pri­mary tumor mass due to altered homotypic adhesion phenomena, (b) the mechanism of blood transportation of metastatic tumor cells by heterotypic cell interactions, and (c) the arrest in organs by specific interactions with the target tis­sue. Genotypic and phenotypic evolution during progression in vivo toward metastasis was de­monstrated using a multiple-drug-marked benign murine tumor cell line by Lagarde et al. (1983).

Motility. In general, increased or excessive cell motility in vivo is a characteristic of malignant cells (Fidler 1974), although more studies have been carried out in experimental systems in vitro than in vivo. The studies of Wood et al. (1954) suggested that the locomotion of tumor cells in vi­vo is nondirectional and lacks chemotaxis, al­though other investigators, including Ozaki et al. (1971), suggested that tumor cells secrete a che­motactic factor which may influence motility and invasiveness. Increased motility in vivo is not unique to tumor cells; it is also a characteristic of normal cells during embryogenesis, regeneration, and wound healing (Fidler 1975). Taptiklis (1968, 1969) intravenously injected dis­sociated cells from cancerous, hyperplastic, and normal thyroid glands. The cells penetrated the endothelial cells of vessels and migrated to extra­vascular tissues. These cells grew in syngeneic thyroxine-deficient mice but not in normal mice. The normal cells survived as long as 1 year and were even induced to become hyperplastic, "a finding that may help to shed light on the phe­nomenon of prolonged dormancy and delayed growth of metastatic tumors in man" (Willis 1973).

Irradiation. X-irradiation of host animals before intravenous injection of tumor cells had been re­ported to increase the incidence of tumor cell im­plantation in the lung (Fidler 1976). Some investi­gators attribute this to host immunosuppression, while others suggest that injury to endothelium of pulmonary vessels leads to increase in trapping of tumor cell emboli. However, Suzuki (1983) found that, while preirradiation of murine hosts en­hanced growth of tumor cells in the lung follow-

Page 164: Respiratory System

154 Bernard Sass and Annabel G. Liebelt

ing intravenous injection, it suppressed lung me­tastasis from tumors transplanted into the muscles of the leg. These findings strongly suggest that experimental and spontaneous systems of metas­tasis do not give the same results. Fisher and Kripke (1977, 1978) and Spellman et al. (1977) showed that pulmonary metastases from ultraviolet-induced tumors developed more readily in ultraviolet-irradiated recipients than in nonirradiated recipients. This enhancement was related to a cellular factor, since it could be trans­ferred by lymphoid cells and partially purified T­lymphocytes from the tumor-bearing animals to normal recipients.

Immune Factors. The immunogenicity of the tu­mor and the immunosurveillance of the host have been implicated in the spread of tumor cells. Ketcham et al. (1966) suggested that the in vivo growth and spread of malignant cells appears to be controlled by some defense mechanism of the host. Crile (1969) proposed that uninvolved reac­tive or hyperplastic regional lymph nodes in hu­man patients with breast cancer may aid in pre­venting metastasis if the tumor is small. Vaage and Pepin (1983) concluded that an immu­nogenic tumor, such as C3H/He mammary can­cer, may act as a source of antigen, which can at­tract a significant number of lymphocytes to function as a temporary accessory lymphoid or­gan, constituting an early and potent source of systemic immune protective factors. This is mani­fest as resistance against vascular dissemination and growth of cancer cells. Sugarbaker and Cohen (1972) and Fogel et al. (1979) demonstrated that primary and metastatic tumors vary antigenically from one another. Ac­cording to Fidler (1980), there have been numer­ous conflicting reports on the effects of spontane­ous or induced tumors in relation to the role of the immune response. Fidler and Kripke (1980) showed that weakly immunogenic tumors metas­tasized more readily in immunocompetent ani­mals than in immunodeficient animals. The growth and metastasis of a highly antigenic tumor was eliminated by immunologic means in an im­munocompetent host. Thus the relationship be­tween immunocompetence and metastasis is far from clear at the present time. Fisher and Kripke (1977, 1978) have demon­strated increased tumor growth in vivo related to an excess of regulatory T cells that suppress anti­tumor immune response. Fidler (1980) postulated that the factors determining whether inhibition or stimulation of the immune response will predomi-

nate are not known, but probably involve such variables as characteristics of the tumor antigen, the mode of antigen presentation (soluble or cell­bound), and the initial site of interaction with host immune cells. Hanna and Schneider (1983) con­cluded that in mice treated with 17-beta-estradiol there was an association between selective inhibi­tion of natural killer-cell-mediated cytotoxicity in vitro and the enhancement of tumor metastasis. They utilized several different mouse tumor cell lines and suggested the possible role of natural killer cells in natural host resistance against hema­togenous tumor dissemination and speculated on an analogous possibility with tumor cells of hu­man origin. Fidler (1974) found that lymphocytes may stimu­late or inhibit tumor growth in experimental me­tastasis, depending on the ratio of lymphocytes to tumor cells. Tumor cells mixed in low ratio with lymphocytes yielded more lung tumor colonies than did tumor cells without the lymphocytes. When they were mixed in a high ratio of lympho­cytes to tumor cells, fewer lung metastases result­ed. Since clumps of four to five tumor cells result in more lung tumor colonies than single cells, Fidler (1980) postulates that tumor-lymphocyte embolus formation is enhanced by injecting tumor cell clumps, thus increasing the likehood of arrest and tumor growth in the lung.

Tumor Cell Interactions with Extracellular Matrix Barriers. Specific tumor cell products may be as­sociated with the progression of benign tumor cells to malignancy. In vitro tumor cells elaborate degradative enzymes such as proteases, which al­low for movement through connective tissue bar­riers in both murine tumors (Liotta et al. 1977, 1979; Recklies et al. 1982; Van Lamsweerde et al. 1983; Henry et al. 1983) and human tumors (Poole et al. 1978; Recklies et al. 1980). Tumor cells can interact with the extracellular ma­trix in at least four ways, as described by Liotta et al. (1983): 1. Attachment to the matrix via specific plasma

membrane receptors, such as the glycoproteins laminin and fibronectin. A tumor cell receptor for laminin was reported for the first time by Rao et al. (1983) on BL6 murine melanoma cells and by Terranova et al. (1983) for human MCF-7 breast cancer cells.

2. Degradation of matrix components associated with invasion by specific hydrolytic enzymes such as collagenolytic enzyme for types IV and V collagen of basement membrane. Immuno-

Page 165: Respiratory System

fluorescence and immunoperoxidase tech­niques used by Barsky et al. (1983) revealed that most invasive carcinomas in humans lacked immuno-reactivity for type IV collagen and laminin of basement membrane, while be­nign and in situ lesions had intact basement membranes.

3. Increased production of matrix components by host cells in response to the presence of the tu­mor.

4. Tumor cell synthesis of matrix components.

Vascular Factors. Disseminated intravascular coagulation is associated with the occurrence of lymphocytic leukemia of man or mice. Small and large vessels may be occluded by thrombi, some of which may contain tumor cells. Widespread deposition of fibrin results from activation of the coagulation system, either by entrance of throm­boplastic substances into the blood or by endo­thelial injury. Intravascular coagulation has a twofold effect on the metastasizing tumor. Firstly, since intravascular coagulation compromises and obstructs blood flow to the small vessels, tumor cell emboli are arrested. Secondly, blood flow to tumor cells already located in the lung parenchy­ma is limited. The ability of actively growing tumors to elicit growth of new capillaries was first observed by Algire and Chalkley (1945). Angiogenesis often initiates the rapid growth (Folkman 1974a), inva­sion, and metastasis of tumors (Folkman 1974b). Greenblatt and Shubik (1968) demonstrated in vitro a not yet purified diffusible factor from tu­mor cells that was mitogenic to vascular endothe­lial cells and stimulated capillaries to prolifer­ate. Gimbrone and Gullino (1976) demonstrated that 30% of mouse mammary HANs elicited neovas­cularization in the eye. The neovascularization is a useful marker for mouse mammary hyperplasia, since it appears before malignant transformation can be detected (Brem et al. 1977). Strum (1983) used chick chorioallantoic-mem­brane-bearing grafts of HANs, plaques, hormone­dependent tumors, and hormone-independent tu­mors from strain GRS to test the ability of these lesions to induce angiogenesis. Fifty percent of the plaques and 63% of the HANs tested were an­giogenic. Eighty percent of the hormone-depen­dent tumors and 97% of the hormone-indepen­dent tumors induced angiogenesis. Study of hormone-independent tumors by elec­tron microscopy showed failure of tumor cells to penetrate basal lamina. Pitelka et al. (1980b), cit-

Metastatic Tumors, Lung, Mouse 155

ed in Strum (1983), stated that metastases may be found in lungs of mice with large tumors of the hormone-independent type. Another possible mechanism by which lymphocytes may enhance tumor growth and metastasis is by promoting vas­cularization. Sidkey and Auerbach (1975, 1976) demonstrated that vascularization in tumor-bear­ing mice could be induced by lymphocytes either reacting against tumors of operating in a graft vs host reaction. Conclusive tests of effects of metas­tatic tumors by lymphocyte-induced angiogenesis have not been done. In summary, the attempts to relate specific char­acteristics with malignancy as measured by the frequency or primary metastasis are not conclu­sive. Correlations with the histological pattern are dependent, at least in spontaneous mammary cancer, on the samples examined. Current infor­mation points to the heterogeneous nature of many neoplasms. Certain tumor characteristics mayor may not be correlated with metastasis, de­pending on more than one characteristic, for in­stance, growth rate and duration. Immunologic, hormonal, and possibly other homeostatic mech­anisms between the host and the tumors play var­ious roles. Finally, the incidence of pulmonary metastasis appears to vary depending on the tu­mor system utilized, that is, whether it is an artifi­cial or spontaneous system.

Comparison with Other Species

Here we briefly compare examples of metastasis to the lung of tumors in domestic animals and several examples of induced tumors metastatic to the lungs of rats.

Occurrence. The single most important and fre­quent tumor metastasizing to the lungs of cats and dogs is osteosarcoma. Nielsen et al. (1954) and Owen (1969) report metastatic rates of 45%-60% for the dog, and Brodey et al. (1963) suggested that the postsurgical metastatic rate in dogs ap­proached 100%. Four of 11 feline osteosarcomas metastasized to the lung (Engle and Brodey 1969). By contrast, chondrosarcoma of the sheep (Sulli­van 1960) and dog (Brodey et al. 1974) tend to grow more slowly and not metastasize. Osteosarcomas of the rat induced by radioiso­topes and chemical carcinogens (dimethylbenzan­thracene, 3,4-benzopyrene, and chelated copper compounds) can be used as models for osteosar­comas in dog and man, since they are similar his­tologically and often metastasize (65% or more)

Page 166: Respiratory System

156 Bernard Sass and Annabel G. Liebelt

(Owen 1966, 1967). Metastasis to the lung of ca­nine mixed mammary tomors occurs frequently, but the rate of metastasis has not been accurately determined. Rats treated with estrogens have been reported to have an 8% rate of metastasis of mammary tumors to the lung (Cutts 1966). This rate of metastasis is lower than that reported for mice. Hepatocellular tumors are uncommon in domes­tic animals; they occur in descending order of fre­quency in the ox, sheep, dog, cat, pig, and horse. Although four out of 13 liver cell tumors of cattle and two out of 21 liver cell tumors of sheep had metastasized, few metastases were found in the lung (Anderson and Sandison 1968; Anderson et al. 1969). Rats fed hepatocarcinogens can serve as a model of lung metastasis. Richardson and Borsos­Nachtnebel (1951) induced hepatocellular carci­noma by 3'-methyl-diaminoazobenene; 74% me­tastasized to the lung.

Ultrastructure. Brooks (1970) observed hyperplas­tic type B (II) cells in the narrow zone of lung tis­sue which surrounded mammary cancer metas­tases in inbred mice (see above). However, in an electron microscopic investigation by Ludatscher et al. (1967) of Morris hepatoma 5123 transplant­ed into strain Buffalo rats, there was no mention of an alveolar cell response to pulmonary metas­tasis. Brooks (1970) also cited three ultrastructural studies oftype B hyperplasia directly related to in­jury of alveolar type A cells in other species. Brooks concludes that the epithelial hyperplasia, in apparent response to the presence of tumors, may depend on the tumor type, the species, and damage to type A cells. Further studies to clarify the nature of this finding will necessitate ultra­structural investigation. The ultrastructural studies of Pitelka et al. (1980b) on metastatic mammary carcinomas in the lungs of mice led to the conclusion that interruptions in the basal lamina of primary and metastatic mam­mary tumor cells are extremely rare. Such inter­ruptions in basal lamina were also seen in chemi­cally induced mammary tumors of rats (Fisher et al. 1975) and mice (Tarin 1969). These findings suggest that interruptions or discontinuities in the basal lamina of mammary tumors provide path­ways for active emigration of malignant cells to the surrounding stroma. Pitelka et al. (1980b) cites several ultrastructural studies of human breast tu­mors in which there were occasional interruptions in the basal lamina in benign lesions and more

frequent and extensive discontinuities or com­plete absence of basal lamina in tumors of in­creasing grades of malignancy. Acknowledgments. We thank Dr. Charles Frith, Intox Laboratories, Little Rock, Arkansas for his contribution of the histological material from which Figs. 184, 185 and 188-195 were obtained, and Mr. Larry Ostby for photomicrography.

References

Algire GH, Chalkley HW (1945) Vascular reactions of nor­mal and malignant tissues in vivo. I. Vascular reactions of mice to wounds and to normal and neoplastic trans­plants. JNCI 6: 73-85

Altevogt P, Fogel M, Cheingsong-Popov R, Dennis J, Ro­binson P, Schirrmacher V (1983) Different patterns of lectin-binding and cell surface sialylation detected on related high- and low-metastatic tumor lines. Cancer Res 43: 5138-5144

Anderson JC, Fugmann RA, Stolfi RL, Martin DS (1974) Metastatic incidence of a spontaneous murine mam­mary adenocarcinoma. Cancer Res 34: 1916-1920

Anderson U, Sandison AT (1968) Tumors of the liver in cattle, sheep and pigs. Cancer 21: 289-301

Anderson U, Sandison AT, Jarrett WFH (1969) A British abattoir survey of tumours in cattle, sheep and pigs. Vet Rec 84: 547-551

Apolant H (1906) Die epithelialen Geschwiilste der Maus. Arb K6niglichen Inst Expt Ther 1 : 7 -62

Ashburn LL (1937) Spontaneous mammary tumors in mice. Factors influencing the incidence of metastases. US Public Health Reports 52: 915-929

Barsky SH, Siegal GP, Jannotta F, Liotta LA (1983) Loss of basement membrane components by invasive tumors but not by their benign counterparts. Lab Invest 49: 140-147

Bashford EF, Murray JA (1904) The zoological distribu­tion, the limitations in the transmissibility, and the com­parative histological cytological characters of malignant new growths. In: Scientific reports, the cancer research fund. Taylor and Francis, London

Borrel A (1903) Epithelioses infectieuses et epitheliomas. Ann de \'Inst Pasteur 17: 81-118

Brem SS, Gullino PM, Medina D (1977) Angiogenesis: a marker for neoplastic transformation of mammary pa­pillary hyperplasia. Science 195: 880-882

Brodey RS, Sauer RM, Medway W (1963) Canine bone neoplasms. J Am Vet Med Assoc 143: 471-495

Brodey RS, Misdorp W, Riser WH, Van der Heul RO (1974) Canine skeletal chondrosarcoma: a clinico­pathologic study of 35 cases. J Am Vet Med Assoc 165: 68-78

Brooks RE (1970) Mouse mammary tumor metastases in lung: an electron microscopic study. Cancer Res 30: 2156-2165

Coman DR (1953) Mechanisms responsible for the origin and distribution of blood-borne tumor metastases: a re­view. Cancer Res 13: 397 -404

Consolandi G, Veronesi U, Briziarelli G (1958) Metastatic spread of mammary cancer: a comparative study in hu-

Page 167: Respiratory System

man beings and mice. In: Severi L (ed) Proceedings of II international symposium on mammary cancer. Division of Cancer Research, Perugia, pp 791-804

Crile G Jr (1969) Possible role of uninvolved regional nodes in preventing metastasis from breast cancer. Can­cer 24: 1283-1285

Cutts JH (1966) Estrogen-induced breast cancer in the rat. Can Cancer Conf 6: 50-68

Dennis JW, Donaghue TP, Kerbel RS (1981) Membrane­associated alterations detected in poorly tumorigenic lectin-resistant variant sublines of a highly malignant and metastatic murine tumor. JNCI 66: 129-139

Dunham U, Stewart HL (1953) A survey of transplantable and transmissible animal tumors. JNCI 13: 1299-1377

Dunn TB (1945) Morphology and histogenesis of mam­mary tumors. In: Moulton FR (ed) A symposium on mammary tumors in mice. AAAS, Smithsonian Insti­tute, Washington DC, pp 13-38

Dunn TB (1953) Morphology of mammary tumors in mice. In: Homburger F, Fishman WH (eds) The physiopathol­ogy of cancer. Hoeber-Harper, New York, chap 8, pp 123-140

Dunn TB (1954) Normal and pathologic anatomy of the reticular tissue in laboratory mice, with classification and discussion of neoplasms. JNCI 14: 1281-1433

Dunn TB, Deringer MK (1968) Reticulum cell neoplasm, type B, or the "Hodgkins-like lesion" of the mouse. JNCI 40: 771-821

Engle GC, Brodey RS (1969) A retrospective study of 395 feline neoplasms. J Am Anim Hosp Assoc 5: 21-31

Epstein SS (1966) Lung as transplant site for malignant tu­mors in rodents. Cancer 19: 454-457

Fawcett DW (1981) The cell, 2nd edn. Saunders, Philadel­phia, p45

Fekete E (1938) A comparative morphological study of the mammary gland in a high and low tumor strain of mice. Am J Pathol14: 557-578 + 3 plates

Fidler IJ (1974) Immune stimulation-inhibition of experi­mental cancer metastasis. Cancer Res 34: 491-498

Fidler IJ (1975) Mechanisms of cancer invasion and metas­tasis. In: Becker FB (ed) Cancer: a comprehensive trea­tise, vol 4, Biology of tumors: surfaces, immunology and comparative pathology. Plenum, New York, chap 3

Fidler IJ (1976) Patterns of tumor cell arrest and develop­ment. In: Weiss L (ed) Fundamental aspects ofmetasta­sis. Elsevier, New York, chap 14

Fidler IJ (1978) Tumor heterogeneity and the biology of cancer invasion and metastasis. Cancer Res 38: 2651-2660

Fidler IJ (1980) Lymphocytes are not only immunocytes. Biomedicine 32: 1-3

Fidler IJ, Kripke ML (1977) Metastasis results from preex­isting variant cells within a malignant tumor. Science 197:893-895

Fidler IJ, Kripke ML (1980) Tumor cell antigenicity, host immunity, and cancer metastases. Cancer Immunol Im­munother 7: 201-205

Fidler I, White R (eds) (1981) Design of models for testing cancer therapeutic agents. Van Nostrand, New York (Litton bionetics workshop series)

Fidler IJ, Gersten DM, Hart IR (1978) The biology of can­cer invasion and metastasis. Adv Cancer Res 28: 149-250

Metastatic Tumors, Lung, Mouse 157

Fisher B, Fisher ER (1967) Metastases of cancer cells. In: Busch H (ed) Methods in cancer research, vol I. Aca­demic, New York, chap 5

Fisher ER, Shoemaker RH, Sabnis A (1975) Relationship of hyperplasia to cancer in 3-methylcholanthrene-in­duced mammary tumorogenesis. Lab Invest 33: 33-42

Fisher MS, Kripke ML (1977) Systemic alteration induced in mice by ultraviolet light irradiation and its relation­ship to ultraviolet carcinogenesis. Proc Natl Acad Sci USA 74: 1688-1692

Fisher MS, Kripke ML (1978) Further studies on the tu­mor-specific suppressor cells induced by ultraviolet ra­diation. J Immunol 121: 1139-1144

Fogel M, Gorelik E, Segal S, Feldman M (1979) Differ­ences in cell surface antigens of tumor metastases and those of the local tumor. JNCI 62: 585-588

Folkman J (1974a) Tumor angiogenesis. Adv Cancer Res 19:331-358

Folkman J (1974b) Tumor angiogenesis factor. Cancer Res 34: 2109-2113

Frith CH, Littlefield NA, Umholtz R (1981) Incidence of pulmonary metastases for various neoplasms in BALBI cStCrlfC3H/Nctr female mice fed N-2-fluorenylaceta­mide. JNCI 66: 703-712

Furth J (1946) Experiments on the spread of neoplastic cells through the respiratory passages. Am J Pathol 22: 1101-1108

Gimbrone MA Jr, Gullino PM (1976) Neovascularization induced by intraocular xenografts of normal, preneo­plastic and neoplastic mouse mammary tissues. JNCI 56:305-318

Gorer PA (1940) The incidence of tumours of the liver and other organs in a pure line of mice (Strong's CBA strain). J. Pathol Bacteriol50: 17-24

Grdina DJ, Peters U, Jones S, Chan E (1978) Separation of cells from a murine fibrosarcoma on the basis of size. II. Differential effects of cell size and age on lung reten­tion and colony formation in normal and precondi­tioned mice. JNCI 61: 215-220

Greenblatt M, Shubik P (1968) Tumor angiogenesis: trans­filter diffusion studies in the hamster by the transparent chamber technique. JNCI 41: 111-124

Haaland M (1911) Spontaneous tumours in mice. 4th Sci rep, Imp cancer res fund. Taylor and Francis, London pp 1-113

Hanna N, Schneider M (1983) Enhancement of tumor metastasis and suppression of natural killer cell activi­ty by beta-estradiol treatment. J Immunol 130: 974-980

Henry N, van Lamsweerde AL, Vaes G (1983) Collagen degradation by metastatic variants of Lewis lung carci­noma: cooperation between tumor cells and macro­phages. Cancer Res 43: 5321-5327

Heppner G, Miller BE (1981) Biological variability of mouse mammary neoplasms. In: Fidler I, White R (eds) Design of models for testing cancer therapeutic agents. Van Nostrand, New York, chap 4

Heppner GH, Dexter DL, DeNucci T, Miller FR, Calabre­si P (1978) Heterogeneity in drug sensitivity among tu­mor cell subpopulations of a single mammary tumor. Cancer Res 38: 3758-3763

Heston WE (1940) Lung tumors and heredity. I. The sus­ceptibility of four inbred strains of mice and their hy-

Page 168: Respiratory System

158 Bernard Sass and Annabel G.Liebelt

brids to tumors induced by subcutaneous injection. JNCI 1: 105-111

Kerbel RS, Dennis JW, Lagarde AE, Frost P (1982) Tumor progression in metastasis: an experimental approach us­ing lectin-resistant tumor variants. Cancer Metastasis Rev 1: 99-140

Ketcham AS, Wexler H, Minton JP (1966) Experimental study of metastases. JAM A 198: 157-164

Kyriazis AP, Koka M, Vesselinovitch SO (1974) Metastatic rate of liver tumors induced by diethylnitrosamine in mice. Cancer Res 34: 2881-2886

Lagarde AE, Donaghue TP, Dennis JW, Kerbel RS (1983) Genotypic and phenotypic evolution of a murine tumor during its progression in vivo toward metastasis. JNCI 71: 183-191

Liebelt AG, Liebelt RA (1967) Transplantation of tumors. In: Busch H (ed) Methods in cancer research, vol 1. Aca­demic, New York, chap 4

Liebelt AG, Bailey C, Fu MM (1981) Mammary cancer in RIll mice - another look. AALAS Publications, no. 103, pp 81-3

Liebelt RA, Liebelt AG, Gulledge AA, Calvert J (1968) Au­toregulation - normal organ and tumor homeostasis. In: The Univ. of Texas MD Anderson Hospital and Tumor Institute at Houston (ed) The proliferation and spread of neoplastic cells. Williams and Wilkins, Baltimore, pp 733-768

Liotta LA, Kleinerman J, Catanzaro P, Rynbrandt 0 (1977) Degradation of basement membrane by murine tumor cells. JNCI 58: 1427-1431

Liotta LA, Abe S, Gehron-Robey P, Martin GR (1979) Preferential digestion of basement membrane collagen by an enzyme derived from a metastatic murine tumor. Proc Natl Acad Sci USA 76: 2268-2272

Liotta LA, Rao CN, Barsky SH (1983) Tumor invasion and the extracellular matrix. Lab Invest 49: 636-649

Ludatscher RM, Luse SA, Suntzeff V (1967) An electron microscopic study of pulmonary tumor emboli from transplantable Morris hepatoma 5123. Cancer Res 27: 1939-1952

Marsh MC (1927) Tumor massage and metastases in mice. J Cancer Res 11: 101-107

Marsh MC (1929) Spontaneous mammary cancer in mice. J Cancer Res 13: 313-339

Miller FR, Heppner GH (1979) Immunologic heterogenei­ty of tumor cell sub-populations from a single mouse mammary tumor. JNCI 63: 1457-1463

Murray JA (1908) Spontaneous cancer in the mouse; his­tology, metastasis, transplantability and the regulation of malignant new growth to spontaneously affected ani­mals. 3rd Sci rpt, imp Cancer Res Fund, London, pp 69-115

National Toxicology Program (1982a) Carcinogenesis bioassay of di(2-ethylhexyl)adipate in F344 rats and B6C3F1 mice. Technical Report Series 212. US Dept HHS, PHS, NIH, Washington DC

National Toxicology Program (1982 b) Carcinogenesis bioassay of di(2-ethylhexyl)phthalate in F344 rats and B6C3F1 mice. Technical Report Series 217.US Dept HHS, PHS, NIH, Washington DC

Nicolson GL, Brunson KW, Fidler IJ (1978) Specificity of arrest, survival and growth of selected metastatic variant cell lines. Cancer Res 38: 4105-4111

Nielsen SW, Schroder JD, Smith DL (1954) The pathology of osteogenic sarcoma in dogs. J Am Vet Med Assoc 124:28-35

Owen LN (1966) Bone tumors in the dog. J S Mr Vet Med Assoc 37: 395-401

Owen LN (1967) Comparative aspects of bone tumours in man and dog. Proc R Soc Med 60: 1309-1310

Owen LN (1969) Bone tumors in man and animals. Butter­worth, London

Ozaki T, Yoshida K, Ushijima K, Hayashi H (1971) Studies on the mechanisms of invasion in cancer. II. In vivo effects of a factor chemotactic for cancer cells. Int J Cancer 7: 93-100

Pitelka DR, Hamamoto ST, Taggart BN (1980 a) Epithelial cell junctions in primary and metastatic mammary tu­mors of mice. Cancer Res 40: 1588-1599

Pitelka DR, Hamamoto ST, Taggart BN (1980b) Basal lamina and tissue recognition in malignant mammary tumors. Cancer Res 40: 1600-1611

Poole AR, Tiltman KJ, Recklies AD, Stoker TAM (1978) Differences in secretion of the proteinase cathepsin B at the edges of human breast carcinomas and fibromas. Nature 273: 545-547

Poste G, Weiss L (1976) Some considerations on cell sur­face alteration in malignancy. In: Weiss L (ed) Funda­mental aspects of metastasis. Elsevier, New York, chap 2

Pybus FC, Miller EW (1934) Hereditary carcinoma of mice; a description of 100 consecutive tumors. Newcas­tle Med J 13-14: 151-169

Rao NC, Barsky SH, Terranova VP, Liotta LA (1983) Iso­lation of a tumor laminin receptor. Biochem Biophys Res Commun 111 : 804-808

Recklies AD, Tiltman KJ, Stoker AM, Poole AR (1980) Se­cretion of proteinases from malignant and nonmalig­nant human breast tissue. Cancer Res 40: 550-556

Recklies AD, Mort JS, Poole AR (1982) Secretion of a thiol proteinase from mouse mammary carcinomas and its characterization. Cancer Res 42: 1026-1032

Reddy JK, Rao MS, Moody DE (1976) Hepatocellular car­cinomas in acatalasemic mice treated with Nafenopin, a hypolipidemic peroxisome proliferator. Cancer Res 36: 1211-1217

Reznik G (1983) Spontaneous and secondary lung tumors in the mouse. In: Reznik-Schuller HM (ed) Compara­tive respiratory tract carcinogenesis, vol 1. Spontaneous respiratory tract carcinogenesis. CRC, Boca Raton, chap 6

Richardson HL, Borsos-Nachtnebel E (1951) Study of Ii v­er tumor development and histologic changes in other organs in rats fed azo dye 3'-methyl-4-dimethylamino­azobenzene. Cancer Res 11: 398-403

Sheldon WG, Curtis MB, Kodell RL, Weed L (1983) Pri­mary harderian gland neoplasms in mice. JNCI 71: 61-68

Sidkey YA, Auerbach R (1975) Lymphocyte induced an­giogenesis: a quantitative and sensitive assay of the graft-vs-host reaction. J Exp Med 141: 1084-1100

Sidkey TA, Auerbach R (1976) Lymphocyte-induced an­giogenesis in tumor bearing mice. Science 192: 1237-1238

Slye M, Holmes HF, Wells HG (1915) Spontaneous tu­mors of the liver, associated frequency with other pri­mary growths (mammary and lung). J Med Res 33: 171-182

Page 169: Respiratory System

Spellman CW, Woodward JG, Daynes RA (1977) Modifi­cation of immunological potential by ultraviolet radia­tion. I. Immune status of short-term UV-irradiated mice. Transplantation 24: 112-119

Squartini F, Bistocchi M (1977) Bioactivity of C3H and RIll mammary tumor viruses in virgin female BALBI c mice. JNCI 58: 1845-1847

Stewart HL (1975) Comparative aspects of certain cancers. In: Becker FB (ed) Cancer, a comprehensive treatise, vol IV. Biology of tumors: surfaces, immunology, com­parative pathology. Plenum, New York, chap 10

Stewart HL, Snell KC, Dunham U, Schlyen SM (1959) Transplantable and transmissible tumors of animals. Am Reg Pathol AFIP, Washington DC (Atlas of tumor pathology, section XII, fascicle 40)

Stewart HL, Dunn TB, Snell KC (1970) Pathology of tu­mors and non-neoplastic proliferative lesions of the lungs of mice. In: Nettesheim P, Hanna MG Jr, Dea­theridge JW (eds) Morphology of experimental respira­tory carcinogenesis. USAEC Division of Technical In­formation Extension, Oak Ridge, pp 161-184 (AEC Symposium series, no 21

Stewart HL, Dunn TB, Snell KC, Deringer MK (1979) Tu­mours of the respiratory tract. In: Turusov VS (ed) Pa­thology of tumours in laboratory animals, vol II. Tu­mours of the mouse. IARC Sci Publ no 23, Lyon, pp251-287

Strum JM (1983) Angiogenic responses elicited from cho­rioallantoic membrane vessels by neoplastic, preneo­plastic, and normal mammary tissues from GR mice. Am J Pathol 111: 282-287

Sugarbaker EV, Cohen AM (1972) Altered antigenicity in spontaneous pulmonary metastases from an antigenic murine sarcoma. Surgery 72: 155-161

Sullivan DJ (1960) Cartilagenous tumors (chondroma and chondrosarcoma) in animals. Am J Vet Res 21: 531-535

Suzuki N (1983) Spontaneous versus artificial lung metas­tasis: discrepant effect of whole-body irradiation of NFSA2ALM and NFSA1SLM tumor systems. JNCI 71:835-839

Takayama S (1968) Induction of transplantable liver tu­mors in DBFl mice after oral administration of N. N-2,7-fluorenylenebisacetamide. JNCI 40: 629-641

Talmadge JE, Starkey JR, Davis WC (1979) Characteristics of induced metastatic variants from a cloned metastati­cally homogeneous cell line. (1979-ICN-UCLA Sympo­sium on cell surfaces and malignancy). Supramol Struct [Suppl] 3: 183

Taptiklis N (1968) Dormancy by dissociated thyroid cells in the lungs of mice. Eur J Cancer 4: 59-66

Taptiklis N (1969) Penetration of the vascular endothelial barrier by non-neoplastic thyroid cells in circulation. Eur J Cancer 5: 445-457

Metastatic Tumors, Lung, Mouse 159

Tarin D (1969) Fine structure of murine mammary tu­mours: the relationship between epithelium and connec­tive tissue in neoplasms induced by various agents. Br J Cancer 23: 417 -425

Terranova VP, Rao CN, Kalebic T, Margulies 1M, Liotta LA (1983) Laminin receptor on human breast carcinoma cells. Proc Natl Acad Sci USA 80: 444-448

Thorpe E, Walker AI (1973) The toxicity of dieldrin (HEOD). II. Comparative long-term oral toxicity studies in mice with dieldrin, DDT, phenobarbitone, B-BHC and Y-BHC. Food Cosmet Toxicolll: 433-442

Turusov VS, Takayama S (1979) Tumours of the liver. In: Turusov VS (ed) Pathology of tumours in laboratory ani­mals, vol II. Tumours of the mouse. IARC Sci Publ no 23, Lyon, pp 193-233

Turusov VS, Day NE, Tomatis L, Gati E, Charles RT (1973 a) Tumors in CF-l mice exposed for six consecu­tive generations to DDT. JNCI 51: 983-997

Turusov VS, Deringer MK, Dunn TB, Stewart HL (1973b) Malignant mouse liver tumors resembling human hepa­toblastomas. JNCI 51: 1689-1695

Vaage J, Pepin KG (1983) Plasma cell infiltrate of the pri­mary tumor as a source of early systemic protection against metastases in mice. JNCI 71: 147-155

van der Valk MA (1981) Survival, tumor incidence and gross pathology in 33 mouse strains. In: Hilgers J, Sluy­ser M (eds) Mammary tumors in the mouse. Elsevier, New York, chap 3

van Lamsweerde AL, Henry N, Vaes G (1983) Metastatic heterogeneity of cells from Lewis lung carcinoma. Can­cer Res 43: 5314-5320

Vesselinovitch SD, Mihailovich N, Rao KVN (1978) Mor­phology and metastatic nature of induced hepatic nodu­lar lesions in C57BL x C3HF1 mice. Cancer Res 38: 2003-2010

Ward JM, Goodman DG, Squire RA, Chu KC, Linhart MS (1979) Neoplastic and nonneoplastic lesions in ag­ing (C57BLl6N x C3H/HeN)Fl (B6C3F1) mice. JNCI 63: 849-854

Wexler H (1966) Accurate identification of experimental pulmonary metastases. JNCI 36: 641-645

Wexler H, Minton JP, Ketcham AS (1965) A comparison of survival time and extent of tumor metastases in mice with transplanted, induced and spontaneous tumors. Cancer 18: 985-994

Williams AC, Silcox LE, Halpert B (1935) Carcinoma of the mammary gland in inbred stock of albino mice. Am J Cancer 24: 823-830

Willis RA (1973) The spread of tumors in the human body, 3rd edn. Butterworths, London

Wood S Jr, Holyoke ED, Clason WPC, Sommers SC, War­ren S (1954) An experimental study of the relationship between tumor size and number of lung metastases. Cancer 7: 437-443

Page 170: Respiratory System

NONNEOPLASTIC LESIONS

Bleomycin-Induced Injury, Mouse: A Model for Pulmonary Fibrosis

Drummond H. Bowden

Gross Appearance

No particular gross features distinguish the devel­oping and established fibrotic lesions induced by bleomycin from other types of injury caused by drugs or other chemicals. Mter a single intrave­nous dose, fibrosis is multifocal and rarely conflu­ent. Diffuse lobular and lobar fibrosis may be ex­pected following the intratracheal insufflation of the drug.

Microscopic Features

The sequential events induced by bleomycin are remarkably similar whatever the route of adminis­tration. What does vary is the speed of the re­sponse; a single intravenous injection of bleomy­cin induces pulmonary fibrosis in about 2 weeks, whereas twice weekly intraperitoneal injections produce fibrosis only after some 4-8 weeks. Di­rect administration of bleomycin through the tra­chea also results in the rapid induction of pul­monary fibrosis. Timing of the morphological events is most precise following a single intrave­nous injection. The following account describes changes in Swiss albino mice given a single intra­venous injection of 120 mg bleomycin per kilo­gram. The earliest detectable alteration is demonstrated by bronchoalveolar lavage. The number of cells in control animals is usually < 20 x 104, almost all of them macrophages. Between 1 and 3 days after bleomycin, the yield is more than doubled and up to 10% are polymorphonuclear leukocytes. In tissue sections of the lung, perivascular edema and endothelial swelling with vacuolation are ob­served at 5 days (Fig. 199). These changes are ob­served earlier in the larger pulmonary vessels than in the microvasculature. In some animals, injury does not progress beyond endothelial swelling with concomitant interstitial edema and egress of inflammatory cells. Such lesions are reversible,

the endothelial cells regenerating with complete restitution of a normal air-blood barrier. The most critical cellular event in the genesis of pulmonary injury is destruction of the thin type I epithelium. Damage to the epithelial barrier, though not directly visible by light microscopy, may be inferred by the exudation of fibrin into the air sacs. If this is massive the animals die; if it is limited or multifocal the animals may survive, but the reparative process usually involves fibrosis. As these changes develop the inflammatory cellu­lar response varies. Initially, cells obtained by la­vage and observed in tissue sections are predomi­nantly macrophagic with a variable percentage of granulocytes; later the cellular profile changes, with an increase in lymphocytes and plasma cells, particularly in the perivascular spaces. As fibrosis develops, macrophages trapped in rigid segments of the lung become vacuolated and some binu­cleate and multinucleate forms are seen (Fig. 200). The evolution of fibrosis is observed in two com­partments of the lung, in fibrin-filled alveoli and in the interstitium. Proliferation of fibroblasts is well established between 10 and 14 days and the laying down of collagen is progressive thereafter. Fibrosis, which is predominantly subpleural and most marked in the perivascular and peribronchi­al spaces, is well established by 2-4 weeks after a single injection of bleomycin. The sequence of reparative events is demon­strated most clearly by autoradiography; endo­thelial regeneration is rapid and results, usually, in complete structural restoration; epithelial re­generation on the other hand is prolonged and is accompanied by the development of abnormal cellular forms. Inappropriate division of intersti­tial fibroblasts is associated with the deposition of collagen and the development of a stiff noncom­pliant lung (Fig. 201). The development of fibrosis following the admin­istration of bleomycin is invariably accompanied by distorted regeneration of alveolar epithelium.

Page 171: Respiratory System

Bleomycin-Induced Injury, Mouse: A Model for Pulmonary Fibrosis 161

Fig. 199 (Above). Lung, mouse. Vascular lesion 7 days after a single intravenous injection of bleomycin. There is endo­thelial vacuolation, subendothelial edema, and a perivas­cular cellular exudate, predominantly mononuclear. Hand E, x 500

Early necrosis of type I cells is followed by divi­sion of type II cells but, instead of differentiating to reconstitute a thin barrier of type I cells, the proliferated cuboidal cells persist, creating tu­bule-like alveoli. The cuboidal cells may exhibit a variety of metaplastic changes; giant forms al­most filling the lumen are observed, together with

(Fig. 200 (Below). Lung, mouse. Alveolar exudate 7 days af­ter a single intravenous injection of bleomycin. The pre­dominant cell is the macrophage; binucleate and multi­nucleate forms are not uncommon. Hand E, x 500

ciliated alveolar cells and squamous differentia­tion with keratin production. Such abnormal ep­ithelial regeneration, though seen following a sin­gle intravenous injection of bleomycin, is more pronounced after regular intraperitoneal injec­tions given over a period of some weeks (Fig. 202).

Page 172: Respiratory System

162 Drummond H. Bowden

Fig.201 (Above). Lung, mouse, after twice weekly intraperi­toneal injections of bleomycin for 4 weeks. Dense bands of subpleural, interstitial collagen distort the normal architec­ture, producing a honeycomb pattern. Silver methenamine, x 350 (reduced by 10%)

Fig.202 (Below). Lung, mouse, 12 weeks after twice weekly intraperitoneal injections of bleomycin. No normal alveoli are seen; many of the alveolar epithelial cells bear cilia. H and E, x 1000 (reduced by 10%)

Page 173: Respiratory System

Fig. 203. Lung, mouse. Small pulmonary blood vessel 2 days after a single intrave­nous injection of bleomycin. There is severe cytoplasmic edema of the endo­thelial cells. Hand E, x 8000 (reduced by 20%)

Fig. 204. Lung, mouse. Alveolus 10 days after a single intravenous injection of bleomycin. Some of the type I cells (EP1) are intact; others (EP1(N)) show focal necrosis allowing the exudation of fibrin (F) from the capillaries (C) into the alveolar lumen (A). TEM, x 6000 (re­duced by 20%)

Fig.205. Lung, mouse. Alveolus (A) 3 weeks after a single intravenous injec­tion of bleomycin. A huge type II (EP2) epithelial cell occupies much of the alveolar lumen. TEM, x 6000 (reduced by 20%)

Bleomycin-Induced Injury, Mouse: A Model for Pulmonary Fibrosis 163

Page 174: Respiratory System

164 Drummond H. Bowden

Ultrastructure

The earliest lesions observed with the electron microscope, endothelial blebbing and subendo­thelial edema, are evident within 2 days of the in­jection of bleomycin (Fig. 203). Necrosis of type I alveolar cells is prominent between 7 and 10 days and this breach of the wall is accompanied by ex­udation of fibrin into the air sacs (Fig. 204). Type II cells and bronchiolar cells show no evi­dence of injury. Regenerative activity is prompt, endothelial cells are rapidly replaced, and by 12 days many of the alveoli are lined by cuboidal cells derived from dividing type II cells. Whereas in some alveoli normal differentiation of type II cell to type I cell occurs, in others cuboidal cells persist and a variety of metaplastic forms are ob­served (Fig. 205). Ultrastructural studies confirm the two compo­nents of the fibrotic response. In the early phase fibroblastic activity is directly related to the fibri­nous exudate which follows necrosis of alveolar cells. These lesions tend to resolve. Fibrosis with­in the interstitium is progressive and almost con­stantly associated with epithelial metaplasia.

Differential Diagnosis

The pulmonary cells most susceptible to airborne or blood-borne injury are the type I epithelial cells and the endothelial cells. The response of these cells to injury by agents as diverse as viruses, oxi­dant gases, and blood-borne drugs is similar, the results being dependent upon the dosage and the duration of the insult. Rapid regeneration of both lining layers is the rule, ensuring restitution of the thin air-blood barrier. Delayed or abnormal re­generation or failure to cover the surface wound is associated frequently with proliferation of inter­stitial fibroblasts and the development of fibrosis. What distinguishes bleomycin-induced injury from other forms of drug-related pulmonary dis­ease is the unusual epithelial response involving delayed and prolonged regeneration with inap­propriate differentiation of the proliferated type II cells. Although similar lesions have been described in mice infected with paramyoxovi­ruses (see page 193), the pattern of abnormal epithelial regeneration associated with interstitial fibrosis is certainly unusual in other forms of drug-induced injury to the lung. The other patho­logical features, such as the early endothelial inju­ry with leakage of plasma constituents and in-

flammatory cells followed by rapid regeneration and restitution of vascular integrity, represent the standard reparative response of the lung.

Biologic Features

Bleomycin is an antibiotic derived from Strepto­myces verticillatus(Umezawa 1974). As a cytotoxic agent it has been particularly useful in the treat­ment of squamous cell carcinoma, testicular tu­mors, and lymphomas. It soon became apparent that bleomycin, introduced largely because it has no major toxic effects on kidney or bone marrow, injures the lung. In all cells, normal or neoplastic, bleomycin is degraded by an enzyme which cleaves carboxyamide groups. The responsiveness of a particular cell type to the drug is related to the activity of this enzyme. Epidermal cells in particu­lar contain low levels of the inactivating enzyme, so that bleomycin is able to reach the nucleus, where it induces fragmentation of DNA with sub­sequent block or derangement in the synthesis of DNA, RNA, and protein. These effects are most pronounced in cells entering the mitotic cycle.

Pathogenesis. The primary location of lesions in the endothelial cells of larger pulmonary vessels is related to the blood-borne delivery of the drug. This is in contrast to injury induced by oxidant gases, where the cells exposed to maximal con­centrations in the capillaries are preferentially in­jured (Adamson and Bowden 1974). The circulating drug enters the pulmonary endo­thelium, and the particular sensitivity of these lin­ing cells is probably related to lack of the inacti­vating enzyme. Endothelial injury facilitates fluid transfer to the interstitium and, if the injury is not severe or widespread, rapid regeneration occurs and the animal recovers. Injury to the type I epi­thelium appears to be a critical event, since dis­ruption of this cellular barrier permits egress of plasma proteins into the alveoli. Such alveolar ex­udates may be phagocytosed by macrophages and cleared or they may become organized by fibro­blasts. Persistence of intraalveolar fibroblastic no­dules is unusual, and it is presumed that they are digested by collagenases and phagocytosed. The response of the type II cell may be the deter­mining factor in the genesis of interstitial fibrosis. In most processes of healing, delay in covering a denuded surface may result in unbridled prolifer­ation of connective tissue cells with productive fibrosis. Bleomycin is rapidly taken up by endo­thelial cells and by type I and type II epithelial

Page 175: Respiratory System

Bleomycin-Induced Injury, Mouse: A Model for Pulmonary Fibrosis 165

Table 15. Pulmonary toxicity of bleomycin

Animal

Mouse (Swiss albino)

Hamster Dog Baboon Pheasant

Route of administration

Intraperitoneal

Intravenous

Intratracheal Intravenous Intramuscular Intravenous

a 1 mg is approximately 1 unit of bleomycin sulfate

cells. The attenuated lining cells on both sides of the air-blood barrier are readily injured, whereas the type II cell, although it takes up the drug, is not obviously affected. The natural response of these cells to necrosis of adjacent type I cells is to divide. The dividing cell, replete with bleomycin, is most vulnerable to DNA injury and the abnor­mal cellular forms observed in the reparative phase may be explained in this way (Adamson and Bowden 1977, 1979). While the precise mechanism for the release of fibroblastic activity is not known, the association between delayed or disturbed epithelial restitu­tion and the occurrence of interstitial fibrosis is established. The cellular response in the lung sug­gests that an immunologic mechanism may be in­volved in the genesis of the lesions. Cell-mediated immunity has been proposed as the likely path­way, but the induction of fibrosis in the thymic­deficient nude mouse indicates that an intact cell­mediated immune system is not essential to the development of pulmonary fibrosis in the mouse. Reported changes in T-Iymphocytes in patients given bleomycin are likely to be secondary reac­tions only (Elson et al. 1977).

Frequency of Pulmonary Lesions. In the Swiss albi­no mouse the frequency and severity of pulmo­nary lesions are dose dependent. A single intrave­nous injection of 120 mg bleomycin per kilogram results in a 50% mortality rate and the surviving animals develop pulmonary fibrosis in 2-4 weeks. Twice weekly injections of 20 mg bleomycin per kilogram kill half of the animals and induce fibro­sis in the survivors in 4-8 weeks. The frequency of metaplastic epithelial changes is greatest in ani­mals receiving intraperitoneal bleomycin for at least 8 weeks. Variability of response in different strains of mice is well recognized, with some being unusually re­sistant to the drug. Following the intratracheal ad-

Dose" Reference

20 mg/kg twice weekly Adamson and Bowden (1974, 1979)

120mg/kg Adamson and Bowden (1977, 1979)

5 units/kg Snider et al. (1978) OAmg/kg Fleischman et al. (1971) 1.5 units/kg twice weekly McCullough et al. (1978) 4.12 mg/kg twice weekly Bedrossian et al. (1977)

ministration of bleomycin, collagen production and deposition is high in C57BlI6 mice, interme­diate in DBAI2 and Swiss mice, and low in BALB mice (Schrier et al. 1983). It is not known if the variation in response is due to differences in the activity of the degradative enzyme in the alveolar cells in these particular strains. A further possibili­ty is suggested by the studies of Walford and Bergmann (1979), who demonstrated a relation­ship between the main histocompatibility com­plex and the ability of cells to repair DNA.

Comparison with Other Species

The pulmonary toxicity of bleomycin varies con­siderably from species to species. In dogs, as little as 0.4 mg/kg induces pulmonary lesions, whereas the oorresponding dose in mice is 20 mg/kg by re­peated intraperitoneal injection and 120 mg/kg given as a single intravenous injection (Table 15). In humans, toxic effects were quite frequent when the drug was first introduced. Later, as dosage was reduced, pulmonary complications were much less frequent. The occasional severe reaction to doses as small as 5 mg/kg suggests that individual susceptibility may be an important parameter in determining the response.

References

Adamson IYR, Bowden DH (1974) The pathogenesis of bleomycin-induced pulmonary fibrosis in mice. Am J Pathol77: 185-198

Adamson IYR, Bowden DH (1977) Origin of ciliated alve­olar epithelial cells in bleomycin-induced lung injury. Am J Pathol87: 569-580

Adamson IYR, Bowden DH (1979) Bleomycin-induced injury and metaplasia of alveolar type 2 cells. Relation­ship of cellular responses to drug presence in the lung. Am J Pathol96: 531-544

Page 176: Respiratory System

166 Yohko Emi and Yoichi Konishi

Bedrossian CWM, Greenberg SD, Yawn DH, O'Neal RM (1977) Experimentally induced bleomycin sulfate pul­monary toxicity. Histopathologic and ultrastructural study in the pheasant. Arch Pathol Lab Med 101: 248-254

Elson N, Szapiel S, Fulmer J, Crystal R (1977) Role of cell­mediated immunity in bleomycin-induced pulmonary fibrosis. Am Rev Respir Dis 115 (Suppl): 54 (abstract)

Fleischman RW, Baker JR, Thompson GR, Schaeppi UH, Ilievsky VR, Cooney DA, Davis RD (1971) Bleomycin­induced interstitial pneumonia in dogs. Thorax 26: 675-682

McCullough B, Schneider S, Greene ND, Johanson WG Jr (1978) Bleomycin-induced lung injury in baboons: alter­ation of cells and immunoglobulins recoverable by bronchoalveolar lavage. Lung 155: 337-358

Schrier DJ, Kunkel RG, Phan SH (1983) The role of strain variation in murine bleomycin-induced pulmonary fi­brosis. Am Rev Respir Dis 127: 63-66

Snider GL, Celli BR, Goldstein RH, O'Brien JJ, Lucey EG (1978) Chronic interstitial pulmonary fibrosis produced in hamsters by endotracheal bleomycin. Am Rev Respir Dis 117: 289-297

Umezawa H (1974) Chemistry and mechanism of action of bleomycin. Fed Proc 33: 2296-2302

Walford RL, Bergmann K (1979) Influence of genes asso­ciated with main histocompatibility complex on deoxy­ribonucleic acid excision repair capacity and bleomycin sensitivity in mouse lymphocytes. Tissue Antigens 14: 336-342

Endogenous Lipid Pneumonia in Female B6C3Fl Mice

Yohko Emi and Yoichi Konishi

Synonyms. Cholesterol pneumonia; foamy cell ularly distributed with no specific localization. pneumonia. The nodules are soft and sharply demarcated

from the slightly red surrounding tissue (Fig. 207).

Gross Appearance

Cutaneous application of methylnaphthalene in acetone to mice (to be described later), is followed by lesions in the lungs (Fig. 206). The pleural sur­face bears multiple white spots and nodules, irreg-

Fig. 206. Gross appearance of lungs, mouse, following cu­taneous application of methylnaphthalene. Note irregular surfaces and multiple white spots and nodules

Microscopic Features

Two lesions are commonly observed. One lesion is characterized by foamy cells and cholesterol

Fig. 207. Cut surface of the lungs, mouse, after fixation

Page 177: Respiratory System

Fig.20S (Above). Endogenous lipid pneumonia, lung of mouse treated with methylnaphthalene. Foamy cells in the alveoli, cholesterol crystals, giant cell reaction, and type II pneumocyte proliferation. Hand E, x 200 (reduced by 15%)

crystals in the alveoli and multinucleated giant cell reaction (Fig. 208). Alveolar walls are slightly thickened and type II pneumocytes are hypertro­phied and increased in number. The second lesion is focal alveolar dilatation adjacent to the first le-

Endogenous Lipid Pneumonia in Female B6C3Fl Mice 167

Fig.209 (Below). Endogenous lipid pneumonia, lung of mouse treated with methyl naphthalene. Note compensato­ry focal dilatation of alveoli adjacent to lesion filled with foamy cells. Hand E, x 40 (reduced by 15%)

sion. This emphysema is probably compensatory due to the first lesion (Fig. 209). Inflammatory cells are only seen in alveolar walls of the sur­rounding lung tissue. Foamy cell accumulation is not observed in organs other than the lung.

Page 178: Respiratory System

168 Yohko Emi and Yoichi Konishi

Differential Diagnosis

Grossly, endogenous lipid pnellmonia resembles the appearance of adenoma of the lung but is dif­ferent in color and consistency. The induced nod­ules seen in the pneumonia are relatively soft and white, not translucent, while adenomas are gray­yellow, translucent, and firm. Histolo.gi~ally, l~~id pneumonia is recognized as a ch~omc mtersht.tal pneumonitis with the presence of mtracellular hp­id deposits, and can be differentiated from other forms of pneumonia. Lipid pneumonia and pul­monary lipidosis have a very similar histological appearance. . . Pulmonary lipidosis is foamy cell accumulatIOn m the alveolar space without interstitial pneumoni­tis. In lipid pneumonia, destruction of foam cells often results in the formation of cholesterol crys­tals accompanied by a giant cell reaction. Exoge­nous lipid pneumonia has a different histological pattern from endogenous lipid pneumonia. The lesions of exogenous lipid pneumonia are focal, whereas those of endogenous lipid pneumonia are diffuse. Exogenous lipid pneumonia is also characterized by granulomatous changes includ­ing flattened macrophages and occasional giant cells.

Biologic Features

Exogenous lipid pneumonia is generally caused by oil entering the trachea and being aspirated. Endogenous lipid pneumonia may occur alone, behind a bronchial obstruction, in association with other inflammatory lesions, or in the absence of apparent cause in the lung. In rats endogenous lipid pneumonia can be induced a~te~ prolon~ed breathing of an atmosphere contammg particu­late antimony trioxide or instillation of similar material intratracheally (Gross et al. 1952). In our studies, methylnaphthalene dissolved in acetone was painted on the shaved skin of the back of female B6C3F1 mice at doses of 29.7 or 118.8 mg/kg body weight, twice a week for life. Control mice received acetone in the same man­ner. Methylnaphthalene is not believed to vapor­ize. Mice which had died or been killed were au­topsied and the principal internal organs and the skin were examined histologically. The lesions observed in the lung were diagnosed as endogenous lipid pneumonia. The frequency of pneumonia observed in the three groups was none out of four (controls), three out of 11 (low dose) (27%), and 31 out of32 (high dose) (97%). It was observed as early as 10 weeks after the start of

the experiment in mice that died. Death peaked at 38 weeks; endogenous lipid pneumonia was re­garded as the cause of death. The experiment was terminated at 61 weeks. Spontaneous endogenous lipid pneumonia has not been reported in labora­tory animals. The results with methylnaphthalene suggest that endogenous lipid pneumonia could result from systemic administration of a chemical agent, al­though additional exposure by oral or pulmonary routes was not avoided by this experiment. Genet­ic factors may also be involved, since familial en­dogenous lipid pneumonia with or without hyper­cholesterolemia occurs in man (Kinoshita et al. 1970). Cholesterol crystals seen in endogenous lipid pneumonia may be formed by the destruc­tion of foamy cells. Foamy cells contain lipids, mainly phospholipids, synthesized by type II pneumocytes. Histologically, hypertrophy and proliferation of type II pneumocytes are predomi­nant features in this type of pneumonia. Type II pneumocytes, therefore, play an important role in the development of endogenous lipid pneumonia.

Comparison of Lesions in Humans

Endogenous lipid pneumonia seen in mice treat­ed with methylnaphthalene is severe and causes death in some animals. The histology of the pneu­monia in mice is quite similar to that described in humans, except that interstitial fibrosis is not ob­served in mice but is seen in human (Waddell et al. 1954; Wright and Heard 1976). The relation between lipid deposition and interstitial pneu­monitis is not known. Endogenous lipid pneumo­nia in mice treated with methylnaphthalene does provide a useful model for studying the ~ech~­nisms of this disease, and may have future Imph­cations for the diagnosis and treatment of the hu­man counterpart.

References

Gross P, Brown JHU, Hatch TF (1952) Experimental en­dogenous lipid pneumonia. Am J Pathol28: 211-218

Kinoshita Y, Ogima I, Saito H, et al. (1970) A case of es­sential familial hyperlipemia with marked intracellular lipid storage. Nippon Naika Gakkai Zasshi 59: 971-979 (in Japanese) .

Waddell WR, Sniffen RC, Whytehead LL (1954) The etIol­ogy of chronic interstitial pneumonitis associated with lipid deposition: an experimental study. J Thorac Surg 28: 134-144

Wright GP, Heard BE (1976) The lungs. In: Sy~~ers W St C (ed) Systemic pathology, 2nd edn. vol 1. LlVlngstone, Edinburgh, pp 373-374

Page 179: Respiratory System

Pulmonary Lipidosis, Rat 169

Pulmonary Lipidosis, Rat

Yohko Emi, Ryuichi Higashiguchi, and Yoichi Konishi

Synonym. Alveolar histiocytosis.

Gross Appearance

The weight of affected lungs (as a percentage of the body weight) does not differ significantly from lungs of control rats. The lungs are normal in color but contain multiple small (ca. 1-2 mm) white spots.

Microscopic Features

Multiple focal lesions are present with alveoli filled with foamy cells (Fig.210). Some of these cells are macrophages with abundant, lipid-laden cytoplasm, but others represent proliferation of type II pneumocytes in the alveolar walls (Fig. 211). In a rare large lesion, cholesterol crys­tals may be present. Interstitial pneumonitis is not observed in the surrounding lung.

Differential Diagnosis

Pulmonary lipidosis can be differentiated from lipid pneumonia. Animals generally do not die of pulmonary lipidosis but may succumb to lipid pneumonia. Grossly, the lesions of pulmonary lip­idosis are small and sometimes difficult to recog­nize. Histologically, granulomatous changes ob­served in exogenous lipid pneumonia are not seen in pulmonary lipidosis. Endogenous lipid pneu­monia is a form of chronic interstitial pneumoni­tis with the presence of foamy macrophages and proliferation of type II pneumocytes; pneumoni­tis is absent in pulmonary lipidosis. Accumulation of foamy cells is more prominent in endogenous

lipid pneumonia and sometimes results in de­struction of alveolar walls. In pulmonary lipido­sis, reactive foamy cells accumulate in the alveoli but the alveolar wall is relatively well preserved and giant cell reaction is seldom seen.

Biologic Features

Serum biochemistry values (Table 16) are elevated for total cholesterol, ester cholesterol, and beta­lipoprotein and decreased for high density lipo­protein-cholesterol in hypophysectomized rats. These abnormal values may be caused by general disturbance of lipid metabolism, resulting in pul­monary lipidosis. Pulmonary lipidosis can be induced by anorectic drugs in laboratory animals (Lullmann-Rauch et al. 1972; Gaton and Wolman 1979); the lesions are reversible when administration of the drug is stopped. Myeloid bodies are commonly seen in foamy cells, suggesting that these cells could have originated from type II pneumocytes. Phospho­lipid accumulation in foamy cells may be related to increased acid phosphatase activity. Pulmonary lipidosis in hypophysectomized rats suggests that hormonal regulation by the hypophysis plays an important role in the development of this disease.

Comparison of Lesions in Humans

Histologically, the lesions are quite similar in ani­mals and man. Epidemiologic studies in Eastern Europe and Russia have shown that pulmonary lipidosis is observed in people who have taken an­orectic drugs (Amor et al. 1971; Homann 1974; Hruban et al. 1973; Mach 1972; Rivier et al. 1972;

Table 16. Serum biochemistry of nonoperated and hypophysectomized rats 55 weeks after the operation

Treatment TC HDL-C EC TL TG PL B-LP NEFA

Nonoperated 89.5± 3.5 13 ±1 20± 1 348.5±35.5 111 ±26 177.5±11.5 648± 2 797.5± 2.5 Hypophysectom- 148.5±13.5 6.5±0.5 137±12 405.5±47.5 91.5±10.5 179.5± 0.5 1105±18 633.0 ± 71.0 ized

TC, Total cholesterol; HDL-C, high-density lipoprotein-cholesterol; EC, ester cholesterol; TL, total lipid; TG, triglyceride; PL, phospholipid; P-LP, beta-lipoprotein; NEFA, nonesterified fatty acid

Page 180: Respiratory System

170 Yohko Emi, Ryuichi Higashiguchi, and Yoichi Konishi

Ugriumova 1976). The anorectic drugs are report­ed to act on feeding centers located in the lateral hypothalamic area and ventromedial hypothala­mic nucleus (Baker 1980). In man, pulmonary hy­pertension frequently accompanies pulmonary lipidosis. Pulmonary lipidosis in hypohysectom­ized rats may be induced by their decreased food

Fig.210 (Above). Pulmonary lipidosis, lung ofhypophysec­tomized rat. Alveoli are filled with foamy cells and adja­cent tissue is intact. Hand E, x 40 (reduced by 15%)

intake, resulting in changes in body lipid compo­sition. Properly balanced intake of lipids may pre­vent the development of pulmonary lipidosis.

Fig. 211 (Below). Pulmonary lipidosis, lung ofhypophysec­tomized rat. Alveoli are filled with lipid-containing foamy cells (a), type II pneumocytes (b), and rare cholesterol crys­tals (c). Hand E, x 200 (reduced by 15%)

Page 181: Respiratory System

References

Amor H, Schwingshacki H, Dienst! F (1971) Primary pul­monary hypertension. Presentation of 8 cases probably caused by use of an anorectic drug. Minerva Med 62: 4623-4631

Baker CE Jr (ed) (1980) Physicians' desk reference. Medi­cal Economic Company, Oradell

Gaton E, Wolman M (1979) Histochemical study on the pathogenesis of chlorocyclizine-induced pulmonary lip­idosis. Histochemistry 63: 203-207

Homann G (1974) Pulmonary hypertension following use of appetite depressants. Med Klin 69: 211

Hruban Z, Slesers A, Aschenbrenner I (1973) Pulmonary

Alveolar Lipoproteinosis, Rat

W.Weller

Synonyms. Alveolar proteinosis; endogenous lip­id pneumonia; multifocal histiocytosis; desquam­ative pneumonia; desquamative interstitial pneu­monia (Brewer et al. 1969; Costello et al. 1975; Gaensler et al. 1966; Gough 1967; Liebow et al. 1965; Shortland et al. 1969; Weller 1976).

~.""

L. '~.

~ " , .. '{ .. ,

." ~ ". "

Alveolar Lipoproteinosis, Rat 171

intraalveolar histiocytosis induced by drugs. Toxicol AppIPharmacoI26:72-85

LuHmann-Rauch R, Reil GH, Rossen E, Seiler KU (1972) The ultrastructure of rat lung changes induced by an an­orectic drug (chlorphentermine). Virchows Arch (Cell Pathol) 11: 167-181

Mach J (1972) Unusual increase in incidence of pulmon­ary hypertension. Vnitr Lek 18: 194-196

Rivier JL, Jaeger M, Reymond C, et al (1972) Primary pul­monary arterial hypertension and appetite depressants. Arch Mal Coeur 65: 787 -796

Ugriumova MO (1976) Anorexic preparations and pri­mary pulmonary hypertension. Ter Arkh 48 (2): 126-131

Gross Appearance

The pleural surface of affected rats is quite typi­cal. In initial stages small, white to grayish, round, elevated foci appear on lung surfaces (Fig. 212). If the involvement is minor, these foci can easily be

I . . ~

Fig.212. Lungs, alveolar lipoproteinosis, 12-month-old male Wi star rats

Page 182: Respiratory System

172 W. Weller

overlooked. If the involvement is more extensive, a tendency toward confluence can be observed. The foci appear to expand and become confluent with greater age. In our test series, two different types of additional changes were observed. In one of these, the lesions were nodular, suggesting tu­mor (Fig.213). The grayish white foci differed in size and were well separated from each other. In confluent lesions (the second type), single foci were not recognizable (Fig. 214); the affected lung surface was gray and slightly elevated in relief.

Fig.213 (Above). Lung, alveolar lipoproteinosis, confluent nodular lesions, 25-month-old Sprague-Dawley rat

Fig.214 (Below). Lung, 23-month-old Wistar rat. Alveolar lipoproteinosis, diffuse lesions

Microscopic Features

The usual types of tissue changes in alveolar lip­oproteinosis are presented in Figs.215 and 216. Initially, the lesions involve alveoli in focal areas (Fig.215) but may become confluent. The alveoli are filled with a moderately dense granular mass, made up essentially of lipid-containing (foamy) macrophages. Increased amounts of protein as well as lipid are present (Beaver et al. 1963; Corrin and King 1969; Flodh et al. 1974; Gross and deTreville 1968; Heppleston 1967; Heppleston et al. 1970; Heppleston and Young 1972; Yang et al. 1966). At higher magnification (Fig. 215), the alveoli are seen to be filled with a partly eosinophilic, partly basophilic, PAS-positive, granular material. In some areas, deposits of cholesterol crystals may be seen, usually engulfed by macrophages. These cells have dense, usually centrally placed nuclei and abundant foamy cytoplasm (Fig. 216). In the center of larger foci, the macrophages are often absent, but some shadowy single-cell struc­tures may be found. In the periphery of these larger foci, macrophages with foamy cytoplasm may be observed in varying numbers. The adja­cent alveolar septa of the lung usually appear to be normal, but in some locations numerous cu­boidal alveolar cells (type II pneumocytes) can be identified. In none of the animals are inflammato­ry infiltrates present in alveolar septa or in peri­bronchial tissue. Most of these foci in animals ex­posed to dust are free of dust and, on the whole, distinctly separate from zones of early fibrosis, which do contain dust particles. Cholesterol granulomas with multinucleated giant cells have been described (Beaver et al. 1963; Corrin and King 1969; Gross and de Tre­ville 1968; Pittermann 1973; Pittermann and Deerberg 1974; Weller et al. 1974, 1978; Yang et al. 1966). They are infrequent, however, and have no recognizable relationship to the degree of alve­olar lipoproteinosis (Fig. 217).

Enzyme Histochemical Findings

In our studies, histochemical investigations em­ployed the folowing enzymes: hydro lases (acid and alkaline phsophatases, nonspecific esterase) and oxidoreductases (DPNH-/TPNH-diaph­orase, alpha-glycerophosphate oxidase, succinic dehydrogenase). In most of the type II pneumocytes, enzyme activ­ity was similar to that demonstrated in the same

Page 183: Respiratory System

cells in histologically normal alveoli. In proliferat­ing type II pneumocytes, increased amounts of hydro lases and oxidoreductases were observed. This indicates elevated metabolic activity at the cellular level and correlates with the electron mi­croscropic findings of increased numbers of os­miophilic lamellar bodies in the cytoplasm. The greatest number of cells (macrophages) in the alveoli contained weak or nonreacting enzymes.

Ultrastructure

The light and electron microscopic findings in alveolar lipoproteinosis have been well described. The findings of the present study are not excep­tional. Different opinions exist, however, on the identity of the cells that fill the alveoli. Certain au­thors (Pittermann 1973), some with reservations (Heppleston et al. 1970; Heppleston and Young 1972), consider the cells in alveoli to be type II pneumocytes. In our study these cells were identi­fied as macrophages. Our interpretation agrees with that of Heppleston et al. (1970). The very large, in general uninuclear, macro phages which fill the alveoli contain relatively regular, large, round inclusions confined by a simple membrane. These inclusions are filled with layers of fine os­miophilic lamellae, often concentrically arranged. The cytoplasm of these cells appears only as a dark, narrow matrix between these lamellar parti­cles. Other organelles have not been identified. The nuclei are primarily eccentrically located, and frequently appear to be indented by the inclusion bodies. The inclusion bodies are often adjacent to the cell membrane (Fig. 218). In addition to these round, intracytoplasmic particles, some cells also contain longitudinal and optically empty hollow spaces from which crystalline, needle-shaped de­posits had been released during fixation and em­bedding. The affected intraalveolar type II pneumocytes maintain their usual structures, although the os­miophilic lamellar bodies in these cells increase in number. Microvilli are present on the surface. Distinct finely granular deposits, in part consist­ing of fine osmiophilic lamellae, are present free

Fig.215 (Above). Lung, rat, alveolar lipoprotein os is. FocalC> lesion with filled alveoli. Hand E, x 25

Fig.216 (Below). Lung, rat, alveolar lipoproteinosis. Small focus of densely packed macrophages with foamy cyto­plasm in alveoli. Methylene blue and basic fuchsin, x 630

Alveolar Lipoproteinosis, Rat 173

Page 184: Respiratory System

174 W. Weller

Fig.217. Lung, rat. Subpleural cholesterol granulomas. H and E, x 25

in the alveolar lumen and sometimes located on the alveolar epithelium. Occasionally, these de­posits have a fine vesicular arrangement. The small residual alveolar lumen between pneumo­cytes and macrophages is in some sections filled with these deposits. In the surrounding regions, focal increase of fibers is evident in the intersti­tium. Interstitial vessels are not changed.

Differential Diagnosis

The histologic features of this disease are quite characteristic and usually provide the basis for precise diagnosis. The alveoli in small or large parts of the lung are filled with large lipid-laden cells. The presence of this lipid gives the cyto­plasm a foamy appearance. The lesion is readily distinguished from infectious granulomas by the absence of other inflammatory exudate. In cryp­tococcosis and pneumocystosis, leukocyte re­sponse also may be minimal, but the organisms may be identified with special stains (Gomori's methenamine silver, Gridley's fungus stain, PAS).

Fig.218. Lung, rat, alveolar lipoproteinosis. Osmiophilic lamellar bodies in alveolar macrophages. TEM, x 5000

Biologic Features

The different terms for the same syndrome are based on the differing interpretations of the fea­tures observed by various investigators. In in­volved alveoli, protein-containing material is found, which is believed to originate (Heppleston 1967) from decomposing macrophages. Surfactant and lamellar bodies may also contribute to the protein volume (Heppleston and Young 1972). The lipid volume in altered alveoli is of particular importance (Beaver et al. 1963; Corrin and King 1969; Flodh et al. 1974; Heppleston et al. 1970). The proportion of cholesterol, triglycerides, and especially phospholipids is distinctly increased. Therefore, we suggest this disease should be named alveolar lipoproteinosis rather than alveo­lar proteinosis. The increased phospholipid pro­portion in alveolar lipoproteinosis suggests the in­teresting possibility of a relationship to the lung surfactant system (Corrin and King 1969; Hep­pleston et al. 1970; Heppleston and Young 1972). This disease may impair or invalidate animal experiments and thus be compared to chronic re-

Page 185: Respiratory System

spiratory disease of conventional rats. On the ba­sis of comparison of lesions in the lungs of both species, alveolar lipoproteinosis in rats may serve as a model for human alveolar lipoproteinosis.

Natural History. These changes in the lungs were first described in germ-free rats (Beaver et al. 1963). Further reports followed relative to its ap­pearance in germ-free (Pittermann and Deerberg 1974) as well as specific-pathogen-free rats (Cor­rin and King 1969; Pittermann 1973; Weller 1977) and conventional rats (Kuhn et al. 1966; Yang et al. 1966).

Morbidity. The morbidity is quite variable and sig­nificantly influenced by the age of the rats (Flodh et al. 1974). The percentage of animals with alveo­lar lipoproteinosis increases as animal groups be­come older (Weller et al. 1978). According to Beaver et al. (1963), the percentage of animals with alveolar lipoproteinosis in different groups may vary between 0 and 97%. In published statis­tics the most frequent postmortem finding was alveolar lipoproteinosis (Pittermann and Deer­berg 1974). Another report referred to frequencies as high as 20% (Yang et al. 1966). In our tests, the frequency in untreated control an­im~ls was as great as 55%. Postmortem findings in ammals aged 12-15 months from different breed­e!s revealed a frequency of alveolar lipoproteino­SIS between 40% and 90%. The earliest occur­rences of the disease, as reported in three studies, were at 6 (Flodh et al. 1974), 11 (Pittermann and Deerberg 1974), and 15 months of age (Yang et al. 1966). Clear statements have not been made con­cerning the relationship between morbidity and strain of origin or sex of the rats: reports are even contradictory (Flodh et al. 1974; Weller et al. 1~78; Yang et al. 1966). Although a comparison WIth completely healthy animals could not be made due to the small number of unaffected rats mortality does not seem to be significantly influ~ enced. Organ weigh~s in affected animals may be slightly or .severely mcreased. In normal rats the lung ~eIghs up to 1.5 g; rats with alveolar lipoproteino­SIS have lungs that weigh up to 4 g; and in dust-ex­posed animals, the lungs may weigh as much as 18 g. In hilar lymph nodes, neither macroscopic nor light microscopic changes are observed and the weight of these lymph nodes is not alter;d.

Etiology. When the alveolar lipoproteinosis syn­?rome ~as fi~st ~escribed (Beaver et al. 1963) dur­l~g an m~esttgatlOn of germ-free rats, a cirrhoge­mc, protem-deficient diet was thought to be the

Alveolar Lipoproteinosis, Rat 175

cause. Subsequent reports postulated a causal in­fluence of dust inhalation (Corrin and King 1969; Gross and deTreville 1968; Heppleston 1967; Heppleston et al. 1970; Heppleston and Young 1972; Jennings et al. 1965). However, when alveo­lar lipoproteinosis also appeared in untreated rats kept under controlled conditions (Pittermann 1973; Pittermann and Deerberg 1974; Weller 1977; Weller et al. 1974; Yang et al. 1966), it was concluded that an independent lung disease was present in these rats (Weller et al. 1974, 1978). The pathogenesis of alveolar lipoproteinosis is not yet clarified. A variety of potential etiologies have been studied. According to Gross and deTreville (1968) and Heppleston and Young (1972), a disturbance of the bronchial purification mechanism, an exces­sive production of alveolar macrophages and a lack or insufficiency of autolytic enzymes can be regarded as the first stage in the pathogenesis. An excessive production of surfactant with an in­creased proportion of phospholipid, together with hyperreactivity of type II pneumocytes (Corrin and King 1969; Heppleston et al. 1970), has also bee~ proposed. This suggestion is supported by the mcreased occurrence of free lamellar bodies in alveoli as well as phagocytized lamellar bodies in alveolar macrophages, a process which may be compared to a merocrine secretion passed from type II pneumocytes into the alveolar lumen (Ha­tasa and Nakamura 1965). A pathological surfactant has also been described (Heppleston and Young 1972). Cholesterol, neu­tral fat, and cell decomposition products are able to deactivate normal surfactant. According to Felts (1965), glucose availability is important to li­~ometabolism in the lung. Furthermore, due to an lllcreased capillary permeability the contents of alveoli have been regarded as transudates (Hep­pleston et al. 1970). In addition, an immunologic disturbance has been suggested (Gray 1973). The implications of the cholesterol granulomas (Beaver et al. 1963; Corrin and King 1969; Pitter­mann 1973; Pittermann and Deerberg 1974; Wel­ler et al. 1974; Yang et al. 1966) need to be clarifi­ed. They are probably related to the increased cholesterol concentration in the abnormal lungs. However, only a small proportion of lungs with alveolar lipoproteinosis also contain cholesterol granulomas. According to Kluge (1959), choles­terol supplied intratracheally cannot be regarded as a cause of progressive cholesterol pneumonia. Flodh et al. (1974), however, found an increased number of foamy cells in lungs of rats that had been fed diets rich in cholesterol or triglyceride.

Page 186: Respiratory System

176 W. Weller

Comparison with Other Species

Alveolar lipoproteinosis of rats is not only of im­portance in experimental animals but may also serve as a model for the human disease. The term "pulmonary alveolar proteinosis" was introduct­ed by Rosen et al. (1958) for a human disease that had been previously unknown or misinterpreted. This disease is characterized by a recurring and/ or progressive accumulation of phospholipid­containing material in the alveoli, but without in­flammation. Patients suffer from an increasing dyspnea combined with cough and loss of weight; nearly one-third of them eventually die of car­diorespiratory insufficiency following a protract­ed illness. Since the first description, many other cases have been reported (Weller et al. 1978). With respect to the comparability of lung changes found in men and in rats, the following can be said: spontaneous alveolar lipoproteinosis in rats and alveolar lipoprotein os is in animals after dust inhalation do not differ significantly by light or electron microscopy or by enzyme histochemistry from that described for human patients. Alveolar lipoproteinosis occurs not only in rats and man, but also in hamsters and guinea pigs (Gross and deTreville 1968).

References

Beaver DL, Ashburn LL, McDaniel EG, Brown ND (1963) Lipid deposits in the lungs of genn-free animals. Arch Pathol 76: 565-570

Brewer DB, Heath D, Asquith P (1969) Electron micros­copy of desquamative interstitial pneumonia. J Pathol 97:317-323

Corrin B, King E (1969) Experimental endogenous lipid pneumonia and silicosis. J Pathol97: 325-330

Costello JF, Moriarty DC, Branthwaite MA, Turner-War­wick M, Corrin B (1975) Diagnosis and management of alveolar proteinosis: the role of electron microscopy. Thorax 30: 121-132

Felts JM (1965) Carbohydrate and lipid metabolism of lung tissue in vitro. Med Thorac 22: 89-99

Flodh H, Magnusson G, Magnusson 0 (1974) Pulmonary foam cells in rats of different age. Z Versuchstierkd 16: 299-312

Gaensler EA, Goff AM, Prowse CM (1966) Desquamative interstitial pneumonia. N Engl J Med 274: 113-128

Gough J (1967) Silicosis and alveolar proteinosis. Br Med J 1: 629

Gray ES (1973) Autoimmunity in childhood pulmonary alveolar proteinosis. Br Med J 4: 296-297 (letter)

Gross P, deTreville RTP (1968) Alveolar proteinosis. Its experimental production in rodents. Arch Pathol 86: 255-261

Hatasa K, Nakamura T (1965) Electron microscopic ob­servations of lung alveolar epithelial cells of nonnal young mice with special reference to fonnation and se­cretion of osmiophilic lamellar bodies. Z Zellforsch 68: 266-277

Heppleston AG (1967) Atypical reaction to inhaled silica. Nature 213: 199

Heppleston AG, Young AE (1972) Alveolar lipo-proteino­sis: an ultrastructural comparison of the experimental and human fonns. J Patholl07: 107-117

Heppleston AG, Wright NA, Stewart JA (1970) Experi­mental alveolar lipo-proteinosis following the inhalta­tion of silica. J Patholl0l: 293-307

Jennings IW, Gresham GA, Howard AN (1965) Pulmo­nary lipidosis in laboratory rats. J Reticuloendothel Soc 2: 130-140

Kluge A (1959) Versuche zur Cholesterinpneumonie. In: Verh. Dtsch. Ges. Path. Fischer, Stuttgart, p 274

Kuhn C, Gyorkey F, Levine BE, Ramirez-Rivera J (1966) Pulmonary alveolar proteinosis. A study using enzyme histochemistry, electron microscopy, and surface ten­sion measurement. Lab Invest 15: 492-509

Liebow AA, Steer A, Billingsley JG (1965) Desquamative interstitial pneumonia. Am J Med 39: 369-404

Pittennann W (1973) Pulmonale Alveolar-Proteinose bei alten SPF HAN: Sprague-Dawley-Ratten. XI. Scientific meeting of the Society for Laboratory Animal Science 9-12 May, Antwerp

Pittennann W, Deerberg F (1974) Erkrankungen bei keim­freien Ratten. Fortbildungslehrgang fUr Versuchstier­kunde, 8-12 October 1973, Berlin. Institut fUr Versuchs­tierkunde und Versuchstierkrankheit, University of Ber­lin

Rosen SH, Castleman B, Liebow AA (1958) Pulmonary alveolar proteinosis. N Engl J Med 258: 1123-1142

Shortland JR, Darke CS, Crane WA (1969) Electron mic­roscopy of desquamative interstitial pneumonia. Thorax 24: 192-208

Weller W (1976) Grundlagen der tierexperimentellen Pneumokonioseforschung. In: WT Ulmer, G Reichel (eds) Pneumokoniosen. Springer, Berlin Heidelberg New York, p 49 (Handbuch der inneren Medizin, vol 4/1)

Weller W (1977) Anthrakosilikose. Tierexperimentelle For­schung. Bergbau-Berufsgenossenschaft, Bochum

Weller W, Haacks H, Guzy J1(, Heine W (1974) Silikose­entwicklung im Intraperitonealtest bei Verwendung von keimfreien, SPF- und konventionellen Ratten. Beitr Sili­koseforsch 26: 265-276

Weller W, Kissler W, Morgenroth K (1978) Alveolar-Lipo­protei nose bei Ratten. Z Versuchstierkd 20: 1-18

Yang YH, Yang CY, Grice HC (1966) Multifocal histiocy­tosis in the lungs of rats. J Pathol Bacteriol 92: 559-561

Page 187: Respiratory System

Bronchiolar/ Alveolar Hyperplasia, Lung, Rat 177

Bronchiolar/ Alveolar Hyperplasia, Lung, Rat

Gary A. Boorman

Synonyms. Alveolar hyperplasia; bronchiolar metaplasia; bronchioalveolar adenomatosis; pul­monary adenomatosis; alveolar epitheliolization.

Gross Appearance

The lesion is generally not visible grossly but may appear as white pinpoint foci from the pleural surface.

Microscopic Features

In microscopic sections, foci of hyperplasia ap­pear as poorly circumscribed areas of increased cellularity in the lung (Fig. 219). The increased cellularity is due to increased numbers of cuboi­dal cells lining the alveoli and often intraluminal cells which are predominantly macrophages (Fig. 220). Important features are the lack of com­pression at the margin (Fig.219) and the persis­tence of normal alveolar architecture within the lesion (Fig. 220). At the margin the hyperplastic cuboidal epithelium extends along the alveolar surface of contiguous alveoli (Fig. 221). As the le­sions progress the lining cells may become multi­layered or form papillary projections into the lu-

mina. The cuboidal cells comprising the hyper­plastic alveolar epithelium usually resemble nor­mal type II pneumocytes with little atypia and few mitotic figures.

Differential Diagnosis

Bronchiolar/alveolar hyperplasia can usually be distinguished from bronchiolar/alveolar adeno­ma. However, with large foci of hyperplasia the distinction becomes more difficult, since morpho­logically there appears to be a gradual progres­sion from focal hyperplasia to adenoma and final­ly to obvious carcinoma with distant metastases. This has created some controversy regarding the appropriate diagnostic criteria to separate these entities. Some of the diagnostic difficulty is due to the lack of significant cytological difference be­tween hyperplasia and bronchiolar/alveolar ad­enoma. Thus one has to rely on structural features and growth patterns to distinguish the two lesions. In contrast to focal hyperplasia, adenomas have a more complex and/or solid pattern in which the original architecture is difficult to discern. The margin of the lesion is also useful. In hyperplasia, the margin is not well defined. Contiguous alveoli are partially covered by cuboidal (hyperplastic)

Fig.219. Focal bronchiolar/alveolar hyperplasia, lung, rat. The pleural surface is not raised, adjacent lung is not com­pressed, and the underlying alveolar pattern can be seen throughout the lesion. Hand E, x 120

Page 188: Respiratory System

178 Gary A. Boonnan

Fig.220 (Left). Bronchiolar/alveolar hyperplasia, lung, rat. Alveolar surfaces are lined by a continuous layer of plump cuboidal cells. Much of the increased cellularity is due to free intraluminal cells, which appear to be pulmonary macrophages. Hand E, x 200

cells and there is no evidence of compression. With bronchiolar/alveolar adenoma, the margin between normal and affected tissue is often quite sharp, with compression of adjacent pulmonary tissue. Inflammatory cells such as intraluminal macrophages and perivascular accumulations of lymphocytes are more commonly associated with areas of hyperplasia.

Biologic Features

Several cell types may contribute to the develop­ment of focal bronchiolar/alveolar hyperplasia in the distal lung of the rat. The alveolar epithelium is comprised of two cell types, the type I and type II pneumocyte. The main lining cell of the alveolus is the type I pneumocyte, which is very

Fig.221 (Right). Bronchiolar/alveolar hyperplasia, lung, rat. At the margin of the lesion the hyperplastic cells ex­tend partially into contiguous alveoli. No compression or distortion of adjacent alveoli; hyperplastic cells lining the alveoli are unifonn with little cellular atypia, are more ba­sophilic, and contain few mitotic figures. The intraluminal erythrocytes are not a usual feature and their presence is felt to be an agonal event. Hand E, x 400

flattened and covers about 90%-95% of the alveo­lar surface (Pinkerton et al. 1982). The remaining cells are type II pneumocytes. The type II cell is cuboidal and its cytoplasm contains osmiophilic lamellar inclusions which are believed to be the source oflung surfactant (Kauffman 1980). Injury to the alveolar epithelium resulting in necrosis of type I cells stimulates the proliferation of type II cells to replace the damaged epithelium (Bocking et al. 1981; Mason et al. 1977; Evans et al. 1975, 1976). Hyperplasia of type II cells is, therefore, a normal response contributing to the repair of the epithelial surface. Cells of the terminal bronchiole also may prolifer­ate and migrate into alveoli in response to lung in­jury (Aso et al. 1976). Cells of the intrapulmonary airways that have regenerative capability and pos­sibly contribute to foci of hyperplasia include

Page 189: Respiratory System

Table 17. Bronchiolar/alveolar hyperplasia in F344 rats

Study typea

Gavage Other

Total

Males

7/249 (2.8%) 4/198 (2.0%)

11/447 (2.5%)

Females

4/248 (1.6%) 31200 (1.5%)

7/448 (1.6%)

a Incidence rates from control groups. Animals in gavage studies received com oil 5 mllkg, five times/week for 104 weeks. Includes data from nine NTP technical re­ports issued through 1983

nonciliated bronchiolar (Clara) cells, Kulchitsky (APUD) cells, and ciliated cells (Jeffery and Reid 1975). Bleomycin exposure causes the appearance of ciliated cells in alveoli (Kauffman 1980), ozone exposure causes focal clusters of nonciliated bronchiolar (Clara) cells (Boorman et al. 1980), and nitrosamine will increase Clara and APUD cells (Kauffman et al. 1979). Thus a variety of cell types have the potential to proliferate and to be found in areas of hyperplasia. The type II pneumocytes have a higher mitotic potential (Dormans 1983) and are felt to be the major cell type found in areas of hyperplasia. It must be cautioned, however, that the role of other cell types has not been thoroughly established. Increased emphasis on toxic lesions in carcino­genesis studies has resulted in better documenta­tion of hyperplastic lesions (Ottolenghi et al. 1975; Pour et al. 1976). In nine recent studies re­ported by the National Toxicology Program, the incidence of bronchiolar/alveolar hyperplasia is quite common in control groups of rats (Table 17). In a recently completed inhalation study of chrys­otile asbestos fibers, bronchiolar/alveolar hyper­plasia was a frequent finding and it may represent an early change in the spectrum of lesions leading to bronchiolar/alveolar carcinoma (McConnell, in preparation). Studies relating specific morpho­logical features to biologic behavior, such as pro­gressive growth following transplantation, remain to be done.

Bronchiolar/ Alveolar Hyperplasia, Lung, Rat 179

References

Aso Y, Yoneda K Kikkawa Y (1976) Morphologic and biochemical study of pulmonary changes induced by bleomycin in mice. Lab Invest 35: 558-568

Bocking A, Mittermayer C, von Deimling a (1981) Ure­thane-induced lung hyperplasia. Lab Invest 44: 138-143

Boorman GA, Schwartz LW, Dungworth DL (1980) Pul­monary effects of prolonged ozone insult in rats. Lab In­vest 43: 108-115

Dormans JA (1983) The ultrastructure of various cell types in the lung of the rat: a survey. Exp Pathol24: 15-33

Evans MJ, Cabral D, Stephens RJ, Freeman G (1975) Transformation of alveolar type II cells to type I cells following exposure to N02. Exp Mol Pathol 22: 142-150

Evans MJ, Johnson LV, Stephens RJ, Freeman G (1976) Cell renewal in the lungs of rats exposed to low levels of ozone. Exp Mol Pathol24: 70-83

Jeffery PK Reid L (1975) New observations of rat airway epithelium: a quantitative and electron microscopic study. J Anat 120: 295-320

Kauffman SL (1980) Cell proliferation in the mammalian lung. Int Rev Exp Pathol22: 131-191

Kauffman SL, Alexander L, Sass L (1979) Histologic and ultrastructural features of the Clara cell adenoma of the mouse lung. Lab Invest 40: 708-716

Mason RJ, Dobbs LG, GreenleafRD, Williams MC (1977) Alveolar type II cells. Fed Proc 36: 2697 -2702

National Toxicology Program (NTP) (1983) Technical re­port series, National Technical Information Service, US Department of Commerce, Springfield

Ottolenghi AD, Haseman JK Payne WW, Falk HL, Mac­Farland HN (1975) Inhalation studies of nickel sulfide in pulmonary carcinogenesis of rats. JNCI 54: 1165-1172

Pinkerton KE, Barry BE, O'Neil 11, Raub JA, Pratt PC, Crapo JD (1982) Morphologic changes in the lung dur­ing the lifespan of Fischer 344 rats. Am J Anat 164: 155-174

Pour P, Stanton MF, Kuschner M, Laskin S, Shabad LM (1976) Tumours of the respiratory tract. In: Turusov VS (ed) Pathology of tumours in laboratory animals, vol 1. Tumours of the rat, part 2. IARC, Lyon, pp 1-40 (Publi­cation no 6)

Page 190: Respiratory System

180 G.E.Dagle and A.P. Wehner

Fly Ash Pneumoconiosis, Hamster

G. E. Dagle and A. P. Wehner

Synonyms. Pneumoconiosis is the generic term for the presence of different dusts in the lung and the resultant lesions. Anthracosis, the condition relat­ed to soot in the lungs, may include fly ash and/or other combustion products of fossil fuels (San­ders et al. 1980).

. -

A B

Gross Appearance

The lungs are diffusely tan to gray, with black fo­ci, typically 1-2 mm in diameter, that tend to be distributed in subpleural areas (Fig. 222). Lung weights and volumes are increased after pro­longed exposure .

Fig. 222 A, B. Gross appearance oflungs from hamsters after 16 months of exposure to A low or B high concentrations of fly ash aerosol

Fig. 223. Interstitial reaction in lung of hamster exposed to fly ash. Hand E, x 486 (reduced by 10%)

Page 191: Respiratory System

Microscopic Features

Dust, generally phagocytosed by alveolar macro­phages, aggregates in alveoli throughout all lobes, with a predilection for peribronchiolar and sub­pleural areas (Fig. 223). An interstitial reaction, composed of thickened alveolar septa with promi­nent alveolar epithelial cells and collagenous stro­ma, is associated with the dust accumulation (Fig. 224). Bronchiolization (see "Bronchiolar/

Fig.224 (Above). Bronchiolization (hyperplasia) (straight arrows) and interstitial reaction (curved arrow) in lung of hamster exposed to fly ash. Hand E, x 120 (reduced by 10%)

Fly Ash Pneumoconiosis, Hamster 181

Alveolar Hyperplasia" p 175), composed of pro­liferation of bronchiolar epithelium into alveolar ducts, is more pronounced in areas of peribron­chiolar dust accumulation (Fig. 225). Vesicular emphysema occasionally occurs.

Ultrastructure

The ultrastructural features of this lesion in ham­sters has not been described.

Fig.225 (Below). Bronchiolization (hyperplasia) in lung of hamster exposed to fly ash. Hand E, x 486 (reduced by 10%)

Page 192: Respiratory System

182 G. E. Dagle and A. P. Wehner

Differential Diagnosis

The lesions are associated with relatively high concentrations of dust in the lung. Definitive di­agnosis would depend upon X-ray or diffraction patterns from individual dust particles. Ferrugi­nous bodies and multinucleated giant cells, as formed with asbestos, are lacking. The lesions al­so lack the interstitial fibrosis and alveolar pro­teinosis associated with silicosis. The epithelial proliferation of alveoli and bronchioles is limited to the immediate area of dust accumulation. Since prolonged exposure to high levels of dust is needed to produce the lesions, dust will always be present in close association with lesions.

Biologic Features

Natural History. The effects of inhaled fly ash were studied in the Syrian golden hamster ex­posed to 70 ~g/l fly ash for 6 hi day, 5 dayslweek for up to 20 weeks (Wehner et al. 1981). Dust de­position increased with time and decreased with recovery. The interstitial reaction increased with time but did not diminish significantly after expo­sure stopped. Bronchiolization also increased with duration of treatment and occasionally pro­gressed even after exposures ceased. Mortality for exposed animals was not higher than for controls (Wehner et al. 1981).

Pathogenesis. Dust is phagocytosed by alveolar macrophages; the release of enzymes from alveo­lar macrophages injures alveolar epithelium. This is followed by compensatory hyperplasia of alve-

olar and brochiolar epithelium and, if injury is se­vere, fibrosis (Sanders et al. 1980). Direct effects of fly ash minerals or absorbed materials on epi­thelium cannot, however, be ruled out. Dust is cleared from the lung by mucociliary escalator or accumulates in mediastinal lymph nodes.

Etiology. Fly ash consists of microcrystals and micro spheroids of varying structure, size, and composition - in general, silicates and oxides of aluminum and other minerals that remain after complete combustion of coal. Various materials from the atmosphere may also adhere to the sur­faces of the particles. Thus it is rather difficult to attribute to any specific component a given toxicl pathogenic effect observed after exposure to fly ash.

Frequency. Lesions observed in hamsters exposed to aerosols of fly ash for 6 hi day for 20 months are quantified in Table 18.

Comparison with Other Species

Anthracosis, including that caused by fly ash, is regarded as an innocuous condition in humans (Spencer 1977). Studies of primates exposed to flay ash, at levels lower than those for hamsters in our study, demonstrated lesions "similar to that seen in any 'nuisance' dust" which were "not con­sidered to be evidence of permanent pathological alteration" (MacFarland et al. 1968). Recently, however, it has been suggested that fly ash parti­cles may act as carriers for toxic materials or may be fibrogenic per se in man (Golden et al. 1982).

Table 18. Histological changes related to fly ash exposure of hamsters for 20 months

Lung change observed Exposure Total no. of No. of hamsters Average severity level hamsters with lesions of lesions' (llg/l3)

Dust accumulation 70 61 61 3.25b

17 62 62 2.29bc

0 55 0

Interstitial reaction 70 61 61 2.23b

17 62 56 1.39bc

0 55 4 1.00

Bronchiolization 70 61 44 1.45b

17 62 29 1.17cd

0 55 11 1.18

a Very slight (or very small amount) = 1, slight (or small amount) = 2, moderate = 3, marked = 4, and severe = 5 b Significantly different from sham-exposed controls, p < 0.01 C Sigificantly different from 70-llg/l exposure level, p < 0.01 d Significantly different from sham-exposed controls, p < 0.05

Page 193: Respiratory System

References

Golden EB, Warnock ML, Hulett LD Jr, Churg AM (1982) Fly ash lung: a new pneumoconiosis? Am Rev Respir Dis 125: 108-112

MacFarland HN, Ulrich CE, Martin A (1968) Chronic ex­posure of cynomolgus monkeys to fly ash. In: Walton WH (ed) Inhaled particles. Unwin, Surrey

Sanders CL, Cross FT, Dagle GE, Mahaffey JA (eds)

Asbestosis, Hamster

G. E. Dagle and A. P. Wehner

Synonym. Asbestos pneumoconiosis.

Gross Appearance

At the higher exposure levels (23 ~g/l), the lungs have a mottled appearance with slightly rusty-tan areas replacing the normal pink coloration. Lung weights tend to increase after prolonged expo­sure.

Microscopic Features

Increased numbers of alveolar macrophages and ferruginous bodies are the principal lesions ob­served at lower exposure levels. The macrophages are diffusely distributed, with some aggregation in areas around respiratory bronchioles (Fig. 226). Those engulfing the ferruginous bodies are gener­ally aggregated in alveoli and frequently form multinucleated giant cells (Fig.227). The ferrugi­nous bodies measure up to 20 ~m long and 2 ~m thick, are frequently curved, stain golden yellow with hematoxylin and eosin, and have a typical beaded appearance due to the hemosiderin depo­sited on the asbestos fibers. Numerous macro­phages contain two or more ferruginous bodies. Portions of the ferruginous bodies frequently ap­pear incompletely phagocytosed. Only rarely are ferruginous bodies seen outside of phagocytes. A mild interstitial fibrosis occurs after 7 months of exposure (Wehner et al. 1975). This occurs in areas with aggregated alveolar macrophages and, at high exposure levels, is associated with lym­phocytes and plasma cells (Fig. 228). Proliferation of alveolar and bronchiolar epithelium and cho-

Asbestosis, Hamster 183

(1980) Pulmonary toxicology of respirable particles. Proceedings: DOE symposium series, CONF-791002. National Technical Information Service, DOE, Oak Ridge

Spencer H (1977) Anthracosis. In: Spencer H (ed) Patholo­gy of the lung, vol 1. Saunders, Philadelphia p411

Wehner AP, Dagle GE, Milliman EM (1981) Chronic in­halation exposure of hamsters to nickel-enriched fly ash. Environ Res 26: 195-216

lesterol clefts occur only at very high exposure levels (Fig.229 and 230). Occasionally, alveoli in these hamsters develop vesicular emphysema.

Ultrastructure

The unique feature of asbestosis is the appearance of ferruginous bodies. Formation of these bodies has been previously described in hamsters by Su­zuki and Churg (1969). Phagocytosis of small fragments of asbestos occurs primarily in alveolar macrophages. This is followed by the appearance of hemosiderin in the cytoplasm, then by intracel­lular transport of iron micelles from hemosiderin granules into the phagosomes with the asbestos fi­bers. Progressive concentration of the iron mi­celles occurs in the vicinity of the fibers. The cen­tral fiber, with its coating of hemosiderin, and the investing membranes of the phagosome are re­garded as essential elements of the ferruginous body (Fig. 213).

Differential Diagnosis

The presence of ferruginous bodies is characteris­tic of asbestosis in hamsters and serves to distin­guish it from inflammatory lesions due to other causes.

Biologic Features

Natural History. Hamsters exposed to asbestos dust (-25 ~g/I) 7 h/day, 5 days/week for 11 months experienced respiratory distress which

Page 194: Respiratory System

184 G.E.Dagle and A.P. Wehner

Fig. 226. Peribronchial distribution of macrophages and interstitial reaction after inhalation of asbestos. Hand E, x 80

forced discontinuation of the exposures (Wehner et al. 1975, 1978). Body weight and survival time were significantly decreased at these high dosage levels.

Pathogenesis. Phagocytosis of inhaled asbestos fi­bers by alveolar macrophages is the initial event. This is followed by an influx of additional mature macrophages into alveoli, from which they are usually cleared by the mucociliary escalator. Those macro phages that have phagocytosed long­er fibers tend to remain in the alveoli longer. Longer fibers are associated with more fibrosis. The macrophages have a significant role in fibro­genesis, perhaps through the regurgitation oflyso­somal enzymes during phagocytosis, but the pathogenesis is unclear (Parkes 1982).

Etiology. Several forms of asbestos, chrysotile, crocidolite, amosite, and anthophylite are all ca­pable of inducing asbestosis. Chrysotile was used to produce the lesions in hamsters described in this report.

Fig. 227. Asbestos body in alveolar macrophage. Hand E, x 512

Frequency. Alveolar macrophages and ferrugi­nous bodies are present in lungs of animals ex­posed to asbestos. Asbestosis develops in all ani­mals exposed to asbestos.

Comparison with Other Species

Asbestosis in humans is characterized as bilateral, diffuse, interstitial pulmonary fibrosis and is found in persons occupationally exposed, thus af­fecting miners, insulators, and shipyard workers. The disease in man results in more severe, diffuse pulmonary fibrosis than is observed experimen­tally in hamsters. Other disorders considered to be related to asbestos exposure in humans, but not documented in hamsters, include hyaline plaques of parietal pleura, carcinoma of the lung, and skin corns (Parkes 1982). Asbestos-induced mesothe­liomas of pleura and peritoneum in hamsters are discussed on page 131. The Stanton hypothesis re­lates longer fiber length to increased carci­nogenicity of asbestos (Harington 1981).

Page 195: Respiratory System

Fig.228. Diffuse granulomatous reaction with numerous asbestos bodies, after exposure to high levels of inhaled asbestos. Hand E, x 320

Asbestos inhalation in rats produces an initial increase in the number of alveolar macrophages, followed by diffuse interstitial pulmonary fibro­sis with prominent hyperplasia of alveolar ep­ithelial cells, several kinds of peripheral lung tu­mors (primarily adenomas, adenocarcinomas, and squamous cell carcinomas), and mesothelio­mas (Wagner et al. 1974). Noteworthy differ­ences in experimental lesions in the rat, com­pared to the hamster, are the absence of ferrugi­nous bodies and the presence of malignant pulmonary tumors.

Asbestosis, Hamster 185

Fig.229 (Above). Proliferation of bronchiolar epithelium associated with granulomatous reaction after exposure to high levels of inhaled asbestos. Hand E, x 320

Fig.230 (Below). Cholesterol clefts associated with granu­lomatous reaction following high levels of inhaled asbes­tos. Hand E, x 125

Page 196: Respiratory System

186 Alexander Kast

Fig.231. Asbestos body after inhalation of an asbestos-cement mixture. TEM, bar = l!lm

References

Harington JS (1981) Fiber carcinogenesis: epidemiologic observations and the Stanton hypothesis. JNCI 67: 977-989

Parkes WR (1982) Occupational lung disorders, 2nd edn. Butterworth,London

Suzuki Y, Churg J (1969) Structure and development of the asbestos body. Am J Pathol55: 79-107

Pulmonary Hair Embolism, Rat

Alexander Kast

Synonyms. Hair fragment emboli in the pulmo­nary vascular system; pulmonary embolism caused by hairs; microembolic pulmonary le­sions; skin emboli in lungs.

Wagner JC, Berry G, Skidmore JW, Timbrell V (1974) The effects of the inhalation of asbestos in rats. Br J Cancer 29:252-269

Wehner AP, Busch RH, Olson RJ, Craig DK (1975) Chronic inhalation of asbestos and cigarette smoke by hamsters. Environ Res 10: 368-383

Wehner AP, Dagle GE, Cannon WC, Buschbom RL (1978) Asbestos cement dust inhalation by hamsters. En­viron Res 17: 367-389

Gross Appearance

Pulmonary hair emboli are not visible grossly. Yamamoto et al. (1982), using a stereomicroscope, have seen the foreign bodies from the cut surface of rat lungs.

Page 197: Respiratory System

Microscopic Features

In histologic sections of lungs, foreign body gran­ulomas are seen in medium-sized pulmonary ar­teries, interalveolar capillaries, and alveoli. In the center of these lesions, fragments of skin, hairs, and broken-off shafts of hair follicles, cut trans­versely, obliquely, or longitudinally, may be found. Identification of the foreign bodies is facilitated by the use of azan-Mallory stain and periodic ac-

Fig.232 (Above). Transverse section of an embolic piece of hair embedded in thrombotic material in a pulmonary artery of a male rat killed after 4 weeks of daily injections into caudal veins. Hand E, x 200 (reduced by 20%)

Pulmonary Hair Embolism, Rat 187

id-Schiff reagent (Yamamoto et al. 1982). Under polarized light this material has the same birefrin­gent character as epidermal keratin (Innes et al. 1958; Tekeli 1974). The hair shafts have bright yel­low dichroism in polarized light and with azan they appear as a red and blue double-ring struc­ture (Schneider and Pappritz 1976). Since the structure of the hair is often unmistakably visible in hematoxylin-and-eosin-stained sections, no special methods are necessary to identify these structures (Figs. 232-234).

Fig.233 (Below). Pigmented hair segment with distinctly visible medulla cells in a thrombotic pulmonary artery of a female rabbit given 28 intravenous injections into ear veins. Hand E, x 200 (reduced by 20%)

Page 198: Respiratory System

188 Alexander Kast

From our experiences, giant cell formation is just beginning on the 3rd day after injection. At this time, the embolic hair particles are surrounded by multinucleated giant cells, fibrin, and leukocytes. At the end of subacute toxicological experiments (usually 28 days) in which material is injected in­travenously, most granulomas are found in alveoli or alveolar capillaries. Less commonly, the hair particles are lodged in parietal thrombi in pulmonary vessels, where they cause partial or complete obturation of the lumen. Endovascular and perivascular inflammatory cell infiltration of vessels containing thrombi has been observed (Tekeli 1974; Yamamoto et al. 1982). Long-standing intravascular obturations become rechanneled in varying degrees in most cases. The granulation tissue often extends into the arterial wall, leading to its destruction and consequent pe­riarterial granuloma (Schneider and Pappritz 1976). It is also striking that hair particles are of­ten found embedded in the wall or in alveoli near to pulmonary arteries (Tsunenari and Kast 1977). This phenomenon has been explained by John­ston et al. (1981) on the basis of the ability of the macrophages to carry the foreign body through the vessel wall to reach the alveolar spaces. Infrequently, some arteries become surrounded by an abscess (Yamamoto et al. 1982). Although the injected parts of the epidermis are naturally populated with bacteria, the thromboemboli very seldom result in bacterial infection. In only six

caudal thrombi or pulmonary emboli found among 1422 rats studied (Table 19) was purulent inflammation present in adjacent tissue. No pul­monary infarction has been observed. Only one exception is present in the literature: areas of hemorrhagic infarction were described in a rabbit experimentally injected with a suspension of hair (Innes et al. 1958).

Differential Diagnosis

Problems in differential diagnosis may arise if no hair particles are detected within a granuloma or if the granulomatous processes are extensive. In such cases drug-specific effects may be simulated. Parasitic nodules are to be differentiated in dogs (Schneider and Pappritz 1976). The virogenic giant cell pneumonia, so-called Hecht's pneumo­nia (Sedlmeier and Schiefer 1971; Giese 1974), may be considered. Nests of foam cells in alveoli of rat lungs are common but seem to be involved in lipid metabolism (Flodh et al. 1974) (see page 169). In none of these lesions are the granu­lomas found in blood vessels. Inhaled foreign bodies, such as particles of hairs or plants, may lead to similar pulmonary granulo­mas, including giant cell formation (Fig. 235). For example, Sahu et al. (1975) have reported giant cells in lungs of mice 30 days after the inhalation of asbestos.

Table 19. Lesions in tail veins and lungs in 1422 Sprague-Dawley rats following daily intravenous injections for 28 days (Kast and Tsunenari 1983)

Organ Finding Males Females

Mean±SDa Total (%)b Mean±SCa Total (%)b

Tail veins Total 7.19±2.69 374 (53.2) 7.79±2.48 405 (56.3) Bleeding 2.12±1.91 110 (15.6) 1.81 ±1.58 94 (13.1) Periphlebitis 2.56±1.88 133 (18.9) 4.27±2.13 222 (30.9) Phlebitis 1.58±1.47 82 (11.7) 2.60±1.91 135 (18.8) Swelling of intima 1.38± 1.29 72 (10.2) 1.27 ± 1.09 66 (9.2) Hair in vessel wall 2.10±1.26 109 (15.5) 2.12±1.40 110 (15.3) Thrombophlebitis 3.13±2.32 163 (23.2) 3.69 ± 2.22 192 (26.7)

With hair particle 1.37 ± 1.34 71 (10.1) 1.40±1.30 73 (10.2)

Lungs Total 3.75±2.46 195 (27.7) 3.58±2.49 186 (25.9) Thromboarteritis 0.56±0.87 29 (4.1) 0.42±0.61 22 (3.1)

With hair particle 0.42 ± 0.70 22 (3.1) 0.27 ± 0.49 14 (1.9) With skin particle 0.08 ± 0.27 4 (0.6) 0.06 ± 0.24 3 (0.4)

Giant cell granulomac 3.35 ± 2.35 174 (24.8) 3.29 ± 2.43 171 (23.8) With hair particle 1.96±1.80 102 (14.5) 1.96±2.10 102 (14.2) With skin particle 0.12±0.38 6 (0.9) 0.06 ± 0.24 3 (0.4)

a Mean number of lesions in each group of 15 rats, excluding animals which did not receive all 28 injections b Total number of males = 703, of females = 719 C In alveolar capillaries or alveoli

Page 199: Respiratory System

Pulmonary Hair Embolism, Rat 189

Fig.234. Pulmonary artery of a beagle with embolic hair trapped in a thrombus. Note the concentric layers of the hair. Azan, x 200 (reduced by 15%). Schneider and Pappritz 1976)

Fig.235. Granuloma with foreign body giant cells phagocytosing inhaled particles of plants in pulmonary alveolus of a 2-year-old female rat. Hand E, x 100 (reduced by 15%)

Many reports have accumulated in the literature about pulmonary embolism and foreign body granulomatosis in man and experimental animals caused by the intravenous injection of particles such as cotton wool fibers, microcrystalline cellu-

lose, cornstarch, talc, rubber, metal or glass, and other materials (Simpson 1922; Easton 1952; Gar­van and Gunner 1964; Zientara and Moore 1970; Bowden 1971; Johnston and Waisman 1971; Johnston et al. 1981; Tomashefski et al. 1981).

Page 200: Respiratory System

190 Alexander Kast

Fig.236. Sections of hair embedded in parietal thrombus in the caudal vein of a male rat. Hand E, x 200 (reduced by 15%)

Biologic Features

Pathogenesis. Intravenous injection into caudal veins of rats and mice, veins of ear of rabbits, and into veins of bigger mammals, such as dog and man, is often followed by embolism of skin parti­cles and hair fragments. These may become part of a thrombus at the site of injection or be caught by the pulmonary filter, phagocytosed, and re­moved by foreign body giant cells within a few weeks. Eventually, the integrity of the affected pulmonary arteries and alveoli is restored. The en­tire process belongs to the so-called "pathology of therapy." In one study, we examined histologic sections of lung and caudal vein from each of 1422 rats that had received 28 daily intravenous injections of various test substances. Lesions in caudal veins were found at the site of injection in more than 50% of the rats (Table 19). These lesions included bleeding into the surrounding tissue, periphlebitis and phlebitis, cushion-like swellings of the intima, and thrombosis. The frequency of hair-containing thrombi does not significantly differ between rats injected intra­venously with various test substances at different dosages and their saline-treated controls. There­fore, controls and treated animals are shown to­gether in Table 19.

The current view is that particles of hair, hair folli­cles, or other bits of skin are punched out by the injecting needle and tranferred through the nee­dle into the wall or lumen of the caudal veins. At this site they may become embedded in thrombot­ic material (Fig.236). If not caught in a thrombus in a vein near the site of injection, hair and skin particles are floated to the lungs, where they are stopped in the pulmonary filter. Innes et al. (1958), who were first to describe this condition, also found a fragment of hair attached to the wall of the right ventricle in one mouse, and in the right atrium of another one. According to Purkiss (1975), the pulmonary capil­laries have a diameter of 7-12 !-Lm, and therefore particles greater than 12!-Lm in diameter (such as cellulose fibers with average diameter of 20.5 !-Lm) would be trapped in the vascular bed of the lung. However, due to the existence of arteriovenous shunts in the lungs, not all of them are so en­trapped. Such shunts should enable the emboli to escape into veins and then into the systemic circu­lation. In our trials (Tsunenari and Kast 1977), on­ly intravenously injected nylon 12 globules less than 10!-Lm in diameter have floated with the blood through the lungs into spleen, liver, and kidneys. In an experimental study of these lesions, we pre­pared a suspension in physiological saline solu-

Page 201: Respiratory System

Fig. 237 (Above). Hair particles in pulmonary alveolus of a male rat 3 days after the intravenous injection of a suspen­sion of hair (for methods see Fig.239). Hand E, x 400 (reduced by 20%)

tion containing about 3000 hair particles per mil­liliter. The hairs were taken from the backs of rats and cut into about 10-j..Lm lengths using a cryostat (Tsunenari and Kast 1977). We injected into cau­dal veins of each of 50 male Sprague-Dawley rats a single dose of 0.25 ml of this rat's hair suspen­sion for each 100 g body weight (low dose). A sec­ond group of 50 males were each given 0.75 mll 100 g body weight (high dose). Ten rats of each group were killed on days 3, 7, 14, 21, and 28 after treatment and examined histologically.

Pulmonary Hair Embolism, Rat 191

Fig. 238 (Below). Giant cells phagocytosing hair particles in an alveolus 7 days after the intravenous injection. Hand E, x 400

The development during the first 3 days of pul­monary granulomas that result from hair emboli after intravenous injection has not been de­scribed. Therefore, a quotation from Johnston et al. (1981) in studies of pulmonary embolism in the rat produced by cotton fibers is of interest: "with­in an hour the fibres become covered by platelets and plasma proteins and neutrophil polymorphs are attracted around them. By 2 hours macro­phages congregate around the emboli and, with accumulation of epitheloid cells, granulomas are

Page 202: Respiratory System

192 Alexander Kast

20 a

18

16

14

12

10

8

6

4

2

o 3 days 28

Fig. 239. Average numbers of hair-phagocytosing giant cells in lungs of rats 3, 7, 14, 21, and 28 days after a single intravenous injection (0.75 ml/l00 g body weight) of a sus­pension containing about 3000 hair particles per milliliter cut at about lO!lm length. Right lung: a, apical lobe; b, car­diac lobe; c, diaphragmatic lobe; and d, azygos lobe; L, left lung. (Tsunenari and Kast 1977)

formed. By 16 hours these have increased in size with the appearance of foreign body giant cells, and reach their maximum size in 3 days." In our experiments, the frequency of embolic hair fragments in lungs was dose-dependent, and most of the fragments were observed in the groups killed 3 days after the injection. The fragments at this time were located in arteries or alveolar cap­illaries with little adjacent leukocytic reaction, in­cluding eosinophils, but in several cases the hair fragments were already surrounded by multinu­cleated giant cells (Fig. 237). Giant cells some­times appeared to originate from the endothelium of the involved capillaries. After 7 days, phagocytosis of the bits of hair is complete (Fig. 238). During the following weeks the granulomas decline in size, the hair particles and giant cells disappear gradually, and finally, after 28 days, the lungs are essentially free of le­sions (Fig.239). No hair particles were found in other organs. This confirms the statement of Teke­Ii (1974), who searched for but did not find kera­tin, fragments of hair, or hair follicles in the ves­sels of other organs.

Frequency. In our study of 1422 rats that had been injected intravenously with various materials in toxicology tests (Table 19), lesions in the lungs were observed in 381 of the rats (27%). These le­sions consisted of giant cell granulomas with pha­gocytosed hair in alveoli, and hair-containing thrombi in pulmonary blood vessels (Fig. 232). No differences were attributable to the sex of the ani-

mals. The frequency is surprisingly high if one considers that only one cross section of about 3 11m thickness from one lobe of the lungs was studied in each animal. Schneider and Pappritz (1976) have pointed out that this frequency in such a small random sample of lung indicates the large number of hair particles that must have been injected in the course of the experiments. Innes et al. (1958) found 22 pulmonary granulo­mas (28%) (six containing foreign bodies) in 80 mice dosed intravenously at different periods from 5 through 20 days. Yamamoto et al. (1982) treated their 165 rats intravenously for 6 months and observed lesions in 28%, including such his­topathologic changes as granuloma (23%), throm­bus (18%), giant cells (10%), and arterial abscess (4%). Tekeli (1974) has seen pulmonary embolism after 7-30 days of treatment in 26 of his 120 rats (22% ). No difference has been demonstrated in the fre­quency of pulmonary embolism between carrier­treated controls and test animals given various doses of injectable solutions (Innes et al. 1958; Liehn and Dahme 1975; Yamamoto et al. 1982; Kast and Tsunenari 1983). The more often the an­imals have been injected, the more numerous are the lesions (Innes et al. 1958); however, reparabili­ty of the lesions has to be taken into considera­tion.

Reparability. In our experiments with the injec­tion of hair suspensions (Tsunenari and Kast 1977), phagocytosis has been observed rapidly progressing to nearly complete removal of the em­bolic hair particles within 4 weeks (Fig. 239). However, in our subacute toxicity studies, pha­gocytosis and removal of hairs inoculated through a needle into caudal veins was not com­plete after 6 weeks of recovery. The number of hair particles remaining in the lungs was signifi­cant (Table 20, Fig. 240). This suggests differences in the condition of immunity in individual rats. However, the effects are similar when different suspensions are used, one containing the rat's own hair and another made up of foreign hairs (Kast and Tsunenari 1983). Possibly, the hair of the back of the rat, used in our experiments, is more easily phagocytosed than the caudal (tail) bristles with their thicker cortex. According to Liehn and Dahme (1975), pulmonary hair emboli in their studies disappear within 8-10 weeks after the final intravenous injection. Contrary to the views of Hottendorf and Hirth (1974) and Schneider and Pappritz (1976), who believe that the risk is minor if the hairs at the in-

Page 203: Respiratory System

Pulmonary Hair Embolism, Rat 193

Fig. 240. Embolic hair fragment distinctly affected by phagocytosis but remaining in lung 6 weeks after the last of 28 daily intravenous injections, male rat. Hand E, x 200 (reduced by 15%)

Table 20. Lesions in tail veins and lungs of90 Sprague-Dawley rats injected intravenously daily for 28 days and allowed to recover for 6 weeks (Kast and Tsunenari 1983)

Organ Finding

Tail veins Total Bleeding Periphlebitis Phlebitis Swelling of intima Hair in vessel wall Thrombophlebitis

With hair Lungs Total

Thromboarteritis With hair particle With skin particle

Giant cell granulomac

With hair particle With skin particle

Males

Mean±SDa

3.60±1.52 o 0.40 ± 0.55 1.00± 1.22 0.40±0.55 1.40±1.34 1.80± 1.79 0.60±0.55 0.80±0.84

} None

0.80±0.84 0.40±0.55 None

Total (%)b

18 (38.3) o (0) 2 (4.3) 5 (10.6) 2 (4.3) 7 (14.9) 9 (19.1) 3 (6.4) 4 (8.5)

4 (8.5) 2 (4.3)

Females

Mean±SDa

3.20±1.92 0 0 0.80±1.30 0.40±0.55 1.40±1.14 1.60± 1.52 0.80±1.10 0.20 ± 0.45

0.20±0.45 0.20 ± 0.45

Total (%)b

16 (37.2) o (0) o (0) 4 (9.3) 2 (4.7) 7 (16.3) 8 (18.6) 4 (9.3) 1 (2.3)

1 (2.3) 1 (2.3)

a Mean number oflesions in each group often rats excluding animals which did not receive all 28 injections b Total number of males =47, of females =43 C In alveolar capillaries or alveoli

jection site are carefully clipped and the injection is carried out with care, we agree with other au­thors that no matter how carefully the intravenous injection is done, pulmonary pathological changes are inevitable results of intravenous in­jection!

Comparison with Other Species

In man, three single observations of skin embo­lism have been published by Hirst and Toyama, (1968), Andrew (1976), and Gilbert and Borchard (1980). The skin particles in these cases were be-

Page 204: Respiratory System

194 Alexander Kast

lieved to have been punched out by puncture of the cephalic vein or heart and carried by the blood to the lungs. A similar but very rare finding is known in human pathology: after amniotic fluid embolism, small lanugo hairs of the human fetus are occasionally found in capillaries of maternal lungs (Scotti 1966; Adamsons et al. 1971; Morgan 1979). According to Schneider and Pappritz (1976), the dermal fibrosis at the point of repeated injections in beagle dogs (cephalic vein) forces many hair roots into deeper skin layers, making them more vulnerable for an excision by the needle and transfer into the puncture channel or the vein's wall. These authors identified hair particles with­in the arteries in ten of their 30 dogs (33%) treated over 4 weeks. In the studies of Hottendorf and Hirth (1974), eight of 67 beagles (12%) had granulomas in small pulmonary arteries. Among 64 Himalayan rabbits treated intravenously over 4 weeks, Kast and Tsunenari (1983) discovered five giant cell granu­lomas, two arterial thrombi (Fig. 233), and one fat cell embolus of the lungs (13%).

References

Adamsons K, Mueller-Heubach E, Myers RE (1971) The innocuousness of amniotic fluid infusion in the preg­nant rhesus monkey. Am J Obstet Gynecol 109: 977-984

Andrew IH (1976) Pulmonary skin embolism: a case report. Pathology 8: 185-187

Bowden DH (1971) The alveolar macrophage. Curr Top Pathol55: 1-36

Easton TW (1952) The role of macrophage movements in the transport and elimination of intravenous thorium dioxide in mice. Am 1 Anat 90: 1-33

F10dh H, Magnusson G, Magnusson 0 (1974) Pulmonary foam cells in rats of different age. Z Versuchstierkd 16: 299-312

Garvan 1M, Gunner BW (1964) The harmful effects of par­ticles in intravenous fluids. Med J Aust 51: 1-6

Giese W (1974) Akute interstitielle Pneumonien. In: Doerr W (ed) Organpathologie, vol 1. Thieme, Stuttgart, chap 3

Gilbert P, Borchard F (1980) Hautembolie der Lunge. Pathologe 1: 161-163

HirstAE, ToyamaM (1968) Skin embolism to the lung. lAMA 205: 54

HottendorfGH, Hirth RS (1974) Lesions of spontaneous subclinical disease in beagle dogs. Vet Pathol 11: 240-258

Innes lRM, Donati El, Yevich PP (1958) Pulmonary le­sions in mice due to fragments of hair, epidermis and ex­traneous matter accidentally injected in toxicity experi­ments. Am 1 Pathol34: 161-167

10hnston WH, Waismanl (1971) Pulmonary com starch granulomas in a drug user. Arch Pathol92: 196-202

10hnston B, Smith P, Heath D (1981) Experimental cotton­fibre pulmonary embolism in the rat. Thorax 36: 910-916

Kast A, Tsunenari Y (1983) Hair embolism in lungs of rat and rabbit caused by intravenous injection. Lab Anim 17: 203-207

Liehn HD, Dahme E (1975) Mikroembolische Lungen­veranderungen bei Versuchstieren nach intravenoser Applikation. Med Int Congr. Proceedings of the Euro­pean Society of Toxicology, Series no 345: 319-323

Morgan M (1979) Amniotic fluid embolism. Anaesthesia 34:20-32

Purkiss R (1975) Effects and distribution of intravenously administered cellulose particles in mice. 1 Pharm Phar­macol 27: 290-292

Sahu AP, Dogra RK, Shanker R, Zaidi S (1975) Fibrogenic response in murine lungs to asbestos. Exp Pathol 11: 21-24

Schneider P, Pappritz G (1976) Hairs causing pulmonary emboli. A rare complication in long-term intravenous studies in dogs. Vet Pathol 13: 394-400

Scotti TM (1966) Disturbance of body water and electro­lytes, and of circulation of blood. In: Anderson W (ed) Pathology (Asian edition). Mosby, St. Louis, chap 4

Sedlmeier H, Schiefer B (1971) Entziindungen der Lunge. In: Dobberstein 1, Pallaske G, Stuenzi H (eds) Hand­buch der speziellen pathologischen Anatomie der Haus­tiere, vol 7. Parey, Berlin

Simpson M (1922) The experimental production of macro­phages in the circulating blood. 1 Med Res 43: 77 -144

Tekeli S (1974) Occurrence of hair-fragment emboli in the pulmonary vascular system of rats. Vet Pathol 11: 482-485

Tomashefski JF Jr, Hirsch CS, lolly PN (1981) Microcrys­talline cellulose pulmonary embolism and granulomato­sis. Arch Pathol Lab Med 105: 89-93

Tsunenari Y, Kast A (1977) Hair embolism in lungs of rat after intravenous injection into caudal veins. Arzneim Forsch 27: 1979-1982

Yamamoto H, Imai S, Okuyama T, Tsubura Y (1982) Pul­monary lesions in rats caused by intravenous injection. Acta Pathol Ipn 32: 741-747

Zientara M, Moore S (1970) Fatal talc embolism in a drug addict. Hum Pathol1: 324-327

Page 205: Respiratory System

LESIONS DUE TO INFECTION

Sendai Virus Infection, Lung, Mouse and Rat

David G. Brownstein

Synonyms. Hemagglutinating virus infection of Japan (HVJ); Japanese hemagglutinating virus in­fection (JHV); hemagglutinating virus infection of the mouse (HVM); parainfluenza virus infec­tion.

Gross Appearance

Mice. Asymptomatic or mildly affected mice usually have no gross pulmonary changes other than increased lung weights, although a few mild­ly affected mice have white lines along the course of bronchi, white foci in lymph nodes, and dual lobes of consolidation. Average increases of 50% have been reported by the 5th day and 100% by the 14th day after experimental infection of Swiss mice (Robinson et al. 1968). Overtly ill and geneti­cally susceptible mice have more dramatic in­creases in lung weights (200%-300%) (Parker et al. 1978). In these cases, one or more lung lobes are plum-colored or contain sharply demarcated plum-colored foci which exude frothy sanguinous fluid when cut. If these mice survive into the 3rd week, consolidated foci are gray. Extrapulmonary changes reflect local and system­ic activation of the immune system and stress. Re­gionallymph nodes enlarge during the 2nd week, a change most obvious in the cervical lymph nodes. These nodes may triple or quadruple in weight. Splenic enlargement (20%-50% in weight) also occurs during the 2nd week. Thymic involu­tion is a stress-related change, the degree being roughly porportional to the degree of lung paren­chymal injury.

Rats. Asymptomatic infections are the rule and follow a pattern similar to that described for asymptomatic or mildly affected mice.

Microscopic Features

Mice. Histopathologic changes in experimental and natural disease can be divided into: acute phase, characterized by degeneration and necro­sis of target epithelium and exudative inflamma­tion; reparative phase, characterized by regenera­tion of damaged target cells and interstitial inflammation; and resolution phase, character­ized by a rapid subsidence of inflammation and repair. During the acute phase, which lasts from 8 to 12 days, there is a descending infection of con­ducting airway epithelium. This frequently ex­tends to proximal type II and, to a lesser degree, type I alveolar epithelium (Brownstein et al. 1981; Parker and Richter 1982; Richter 1970, 1973; Zurcher et al. 1977). The result is an acute endo­bronchitis-bronchiolitis and bronchogenic alveo­litis. The earliest airway changes are segmental. Infected bronchiolar epithelial cells are hypertro­phied with eosinophilic foamy, granular, or ho­mogeneous cytoplasm. The nuclei of infected cells are poorly polarized and may be enlarged and vesicular. Inflammatory infiltrates appear shortly after these initial cytological changes (Fig. 241). The lamina propria and adventitia are expanded by edema, dilated lymphatics, and cel­lular infiltrates. Initially, these infiltrates are a mixture of polymorphonuclear leukocytes, reac­tive lymphoid cells with moderate amounts of ho­mogeneous amphophilic or eosinophilic cyto­plasm, and large round or fusiform lymphoreticu­lar cells. The relative number of each type depends on the age and genetic composition of the host and the dose and route (intranasal versus aerosol) of viral exposure. In genetically suscepti­ble or immature mice, neutrophils often predomi­nate as they do after exposure to large doses of vi­rus. Peribronchiolar pulmonary arterioles and venules have increased intimal cellularity. The en­dothelium is hypertrophied and elevated by asymmetric accumulations of leukocytes in the

Page 206: Respiratory System

196 David G. Brownstein

Fig. 241 (Above). Sendai virus infection, acute phase. Bron­chiolar epithelium is hypertrophied with some loss of nu­clear polarity. The adventitia is edematous and infiltrated with lymphoid cells. Hand E, x 354 (reduced by 15%)

subendothelial layer of the intima. There is strik­ing leukocyte pavementing. The adventitia of these vessels is also hypercellular due to leukocyt­ic infiltrates similar to those infiltrating airways. The adventitia is also expanded by edema. Cyto­pathic changes in bronchiolar epithelium are af­forded some specificity if syncytia, cytoplasmic

Fig.242 (Below). Sendai virus infection, acute phase. Bron­chiole with multiple epithelial syncytia. Hand E, x 791 (reduced by 15%)

inclusions, or intranuclear inclusions are present. The lesions may depend, in part, on virus and mouse strain. These changes are usually not seen or are equiv­ocal, since infected cells tend to slough prior to their development. Syncytia usually appear as central clusters of condensed or pyknotic epithe-

Page 207: Respiratory System

Fig. 243 (Above). Sendai virus infection, acute phase. Alve­oli radiating from an infected terminal bronchiole contain a mixed inflammatory exudate. Alveolar septa are edema­tous and infiltrated with mononuclear cells. Hand E, x 345 (reduced by 15%)

lial nuclei surrounded by foamy eosinophilic cytoplasm that crowds adjacent cells (Fig. 242). Cytoplasmic inclusions represent aggregates of redundant nucleocapsids within the cytoplasmic matrix (see Ultrastructure). They are most obvi­ous as homogeneous spherical or irregular eo-

Sendai Virus Infection, Lung, Mouse and Rat 197

Fig.244 (Below). Sendai virus infection, reparative phase. Bronchiole with disorganized regenerating epithelium. Nuclei are pleomorphic and vesicular with prominent nu­cleoli. Hand E, x 245 (reduced by 15%)

sinophilic cytoplasmic bodies surrounded by a narrow halo. This halo is a fixation shrinkage artifact. Intranuclear inclusions are rare in Sen­dai virus infections but intranuclear viral parti­cles have been described in DBAI2 mice (Rich­ter 1970) and intranuclear inclusions have been

Page 208: Respiratory System

198 David G.Brownstein

reported in athymic nude mice (Ward et al. 1976). Near the end ofthe acute phase, infected epitheli­um has sloughed in sheets or as individual cells which lie within the lumen along with an inflam­matory exudate. Because of the segmental nature of the infection, some airways may be devoid of lining cells while others are lined by intact, hyper­trophied epithelium. Airways with desquamating epithelium frequently have focally dense lym­phoid aggregates within the lamina propria, which cause overlying epithelium to bulge into the lumen. The alveolar component of the acute phase is characterized by accumulations of polymorpho­nuclear leukocytes, macrophages, lymphoid cells, and desquamated pneumocytes within alveolar spaces. Alveolar septa are thickened by edema, congested capillaries, and hypertrophied alveolar comer cells. These changes are multifocal, being oriented around infected terminal bronchioles. In severe cases foci of parenchymal inflammation may become confluent, resulting in lobar pneu­monia. Alveolar spaces may fill with fibrin or ex­travasated blood. Foci of emphysema may occur where septal integrity has been destroyed. Near the end of the acute phase fibrin deposits have condensed along denuded alveolar surfaces or as discrete deposits in alveolar spaces. Increased cel­lularity of thickened alveolar septa is due to accu­mulating mononuclear cells (Fig. 243). Inflamed alveoli may be partially atelectatic. The reparative phase is heralded by the appear­ance of regenerating epithelium. This may be seen as early as the 3rd day, but does not predominate over degenerative airway changes until days 8-12. Initially, partially denuded airways contain con­tinuous or interrupted patches of low cuboidal, polygonal, or squamous cells with basophilic cy­toplasm and large pleomorphic vesicular nuclei which are poorly polarized and contain promi­nent nucleoli (Fig.244). Mitotic figures are com­mon in these cells. Once denuded airways are reepithelialized, lining cells become columnar and rapidly assume a normal mucociliary appear­ance. Sessile or pedunculated airway excres­cences are extremely common during the repara­tive phase. These may be composed solely of epithelium or have cores of lymphoreticular cells or fibroblasts. These are ephemeral structures and are rarely seen after the 3rd week. It is common during this phase for terminal bronchioles to be lined by nonkeratinizing stratified squamous epi­thelium. The metaplastic epithelial cells most closely resemble spinous cells, although intercel-

lular bridges are usually difficult to identify. The adventitia and lamina propria of segments under­going repair are less edematous than in the acute phase, and are densely infiltrated with lympho­cytes and plasma cells. The intimal cellularity of pulmonary arterioles and venules seen during the acute phase in usually absent in the reparative phase, but adventitial infiltrates of lymphocytes and plasma cells are prominent. During the reparative phase, alveolar inflamma­tion has shifted from the airspaces to the intersti­tium. Two forms of alveolar repair are recognized. In the first, septa are lined by cuboidal epithelium (adenomatous hyperplasia, alveolar bronchioliza­tion, alveolar epithelialization) (Fig. 245). These epithelial cells are initially undifferentiated, but soon differentiate into pneumocytes or ciliated, mucous or Clara cells. In the second form of re­pair, sheets and nests of metaplastic squamous ep­ithelium fill alveolar spaces (Fig. 246). There is partial or total atelectasis of affected alveoli dur­ing this phase. If fibrin has been deposited it un­dergoes lysis or organization. Macrophages sur­round and infiltrate large fibrin deposits. Plump fibroblasts may also traverse these deposits. Dur­ing the 3rd week collagen bundles can be identi­fied in organizing fibrin depositis. The reparative phase is usually complete by the end of the 3rd week. The resolution phase may be as short as 1 week, so that by the end of the 4th week residual lesions are difficult to identify. There are, however, sever­al changes which may persist for long periods. The most severe sequela, and one that may persist for the life of the animal, is organizing alveolitis with or without bronchiolitis fibrosa obliterans (Fig. 247). This change is the end result of the or­ganization of extensive fibrin deposits in denuded alveoli and terminal airways. Sheets of haphaz­ardly arranged collagen bundles and fibroblasts in natural infections may obliterate parenchymal architecture or may fill alveolar spaces, leaving parenchymal architecture intact. Other residual lesions include foci of emphysema containing in­spissated secretions, cholesterol crystals, and macrophages; focal aggregates of foamy, alveolar macrophages; focal thickening of alveolar septa; focal adenomatous hyperplasia of alveoli; and perivascular and peribronchiolar lymphoplasma­cytic infiltrates. These changes have been identi­fied in mice up to 1 year after infection with Sen­dai virus (Appel et al. 1971; Parker and Richter 1982; Robinson et al. 1968). Athymic nude mice, unlike euthymic mice, devel­op progressive lung changes due to persistent in-

Page 209: Respiratory System

Fig.245 (Above). Sendai virus infection, reparative phase. Alveoli are lined by cuboidal epithelium and septa are in­filtrated with lymphoid cells. Hand E, x 600 (reduced by 15%)

fection with Sendai virus (Iwai et al. 1979; Ward et al. 1976). Pulmonary changes are most often diffuse due to extensive parenchymal involve­ment. Alveolar septa are thickened by edema, neutrophils, and macrophages. Alveolar epithe­lialization is striking and squamous metaplasia is prominent in some cases. Bronchiolar epithelium

Sendai Virus Infection, Lung, Mouse and Rat 199

Fig.246 (Below). Sendai virus infection, reparative phase. Alveolar squamous metaplasia. Hand E, x 600 (reduced by 15%)

is hyperplastic with numerous mitotic figures. Airways may be filled with neutrophils. Lym­phoid infiltrates are usually sparse.

Rats. Information on the microscopic appearance of Sendai-virus-induced lung lesions in rats is lim­ited. Lesions in a colony of mixed rat strains that

Page 210: Respiratory System

200 David G. Brownstein

Fig. 247. Sendai virus infection, resolution phase. Organizing alveolitis. Hand E, x 244 (reduced by 15%)

develop antibodies to Sendai virus have been re­ported but virus isolation was not attempted (Bu­rek et al. 1977). These rats had mild to severe peri­bronchial and perivascular cuffing by lympho­cytes and plasma cells, and interstitial pneumo­nia. Bronchiolar epithelium was hyperplastic and focally ulcerated. Unfortunately, the rats also de­veloped antibodies to pneumonia virus of mice. In the author's experience this virus is capable of producing inflammatory lung disease in the rat. Necrotizing bronchitis has been reported in germ­free rats inoculated intranasally with Sendai virus (Jacoby et al. 1979).

Ultrastructure

Limited ultrastructural pathology has been re­ported of experimental and spontaneous Sendai virus infections in euthymic mice (Brownstein et al. 1981; Parker and Richter 1982; Richter 1970, 1973; Zurcher et al. 1977). The ultrastructure of naturally infected athymic nude mice has also been reported (Ward et al. 1976). The primary site of replication is the epithelium of bronchi and bronchioles and it is within ciliated and Clara cells that ultrastructural evidence of replication is most easily seen. Nucleocapsid assembly occurs in the cytosol at a rate far in excess of the rate of virus assembly. The result is massive accumula-

tions of nucleocapsids, which appear as poorly delineated aggregates or crystalline arrays of rigid hollow fibrils 16-18 nm in diameter. Intranuclear aggregates of similar-appearing fibrils have been reported and these apparently correspond to the intranuclear inclusions seen by light microscopy. The significance of these aggregates is not known; they are rare and appear late in infection, suggest­ing that they may be transported from sites of syn­thesis in the cytosol (Choppin and Compans 1975). Nucleocapsids align beneath modified seg­ments of plasma membrane to initiate exotrophy. These modified segments are thickened by gly­coprotein surface spikes and an electron-dense layer on the inside of the plasma membrane. Bud­ding forms may be spherical, filamentous, or ple­omorphic (Darlington et al. 1970). This appearance of productively infected cells is also seen in infected type II alveolar epithelium (Brownstein et al. 1981). An additional cytoplas­mic inclusion consisting of membrane-bound, randomly arranged hollow fibrils, 15-20 nm in di­ameter, surrounded by 40-nm electron-dense cuffs has been reported in these cells (Zurcher et al. 1977). Incomplete reports indicate viral com­ponents in type I alveolar epithelium and septal capillary endothelium (Parker and Richter 1982; Ward et al. 1976). Only intranuclear nucleocap­sids have been described in the former (nude mice). Viral assembly has not been observed in ei-

Page 211: Respiratory System

ther cell type. Both of these may be examples of abortive infections. Alveolar macrophages are susceptible to abor­tive infections in vitro (Eustatia et al. 1972; Mims and Murphy 1973), but ultrastructural evi­dence of viral replication is lacking. The author has seen numerous virions and nucleocapsids within heterophagosomes of alveolar macro­phages in experimentally infected mice. This perhaps represents phagocytosis of debris con­taining viral particles.

Differential Diagnosis

Mice. Sendai virus pneumonia must be distin­guished from pneumonias caused by mouse co­ronaviruses, K virus, pneumonia virus, Mycoplas­ma pulmonis, and Corynebacterium kutscheri. The chronic wasting disease produced in athymic nude mice must be distinguished from similar syndromes caused by mouse coronarviruses, mouse adenovirus, Pneumocystis, Giardia, Spiro­nucleus, and Toxoplasma.

Rats. Sendai virus pneumonia must be distin­guished from pneumonias caused by rat coronavi­rus, pneumonia virus, Mycoplasma pulmonis, Streptococcus pneumoniae, and Corynebacterium kutscheri. Some pulmonary changes caused by Sendai virus infection mimic those changes caused by expo­sure of rodents to halogenated aromatic hydro­carbons (Reid et al. 1973), oxidant gases (Ste­phens et al. 1974), and other toxicants which have the terminal conducting airways as target struc­tures.

Biologic Features

Natural History. Sendai virus causes acute limited infections in immunocompetent rodents. There is no evidence for latency or chronic infections (Fu­jiwara et al. 1976; van der Veen et al. 1974). En­zootic and epizootic forms exist. Enzootic infec­tions occur in partially immune rodent colonies where susceptible individuals are regularly intro­duced to perpetuate the infection. This can occur in breeding or open colonies. In breeding colo­nies, the susceptible population is the weaning age animals (3-6 weeks), due to their declining passive immunity (Parker and Reynolds 1968). Enzootic infections are usually subclinical.

Sendai Virus Infection, Lung, Mouse and Rat 201

Epizootic infections occur in naive rodent colo­nies and either die out after 2-7 months or be­come enzootic if the proper conditions exist (Parker and Reynolds 1968; Parker et al. 1978). Clinical signs may be associated with epizootic in­fections in mice but have not been reported in rats. Such factors as strain susceptibility, age, hus­bandry, shipping, and copathogens are important in precipitating overt disease in mice (Jakab 1974; Jakab and Dick 1973; Parker et al. 1978; Ward 1974; Zurcher et al. 1977). Breeding colonies may exhibit neonatal or weanling mortality, prolonged gestation, fetal resorption, or runting in young mice (Bhatt and Jonas 1974; Iwai et al. 1979). Adult mice may exhibit anorexia, depression, ruf­fled fur, hunched posture, chattering, conjunctivi­tis, and photophobia. Sendai virus is transmitted by aerosol and contact routes. Naso- and oropharyngeal secretions de­velop high infectivity titers during the 1st week. Fifty to seventy percent of mice will become in­fected after 24 h of contact with a transmitter mouse (van der Veen et al. 1970, 1972). Attack rates are lower for aerosol transmission unless multiple transmitters are present (van der Veen et al. 1974). Athymic nude mice infected with Sendai virus are persistently infected. Most exhibit dramatic weight loss, depression, wrinkled skin, dyspnea, and cyanosis and usually die between 2 and 10 weeks later (Iwai et al. 1979; Ward et al. 1976).

Pathogenesis. Viral replication is restricted to the respiratory tract in natural infections, although a low-level transient viremia may occur. Peak titers are reached in 3-6 days and virus is usually not recoverable after 8-12 days (Appel et al. 1971; Parker et al. 1978; Robinson et al. 1968; Sawicki 1962; Stewart and Tucker 1978; van Nunen and van der Veen 1967). Adult mice eliminate virus earlier than suckling mice and develop lower peak titers (Sawicki 1961). Outbred mice eliminate vi­rus earlier than inbred mice (Stewart and Tucker 1978). Seroconversion (hemagglutination inhibi­tion, complement fixation) is usually detectable on days 7-9 (Appel et al. 1971; Robinson et al. 1968; Stewart and Tucker 1978; van Nunen and van der Veen 1967). Mice are usually leukopenic by the 7th day of infection; leukocytosis follows on days 8-11 (Robinson et al. 1968). Offspring of naturally infected dams rapidly acquire neutraliz­ing and complement-fixing antibodies of the IgG1

and IgG2 subclasses with the onset of nursing. These titers plateau on days 7 -14 and then rapidly decline (lida et al. 1973).

Page 212: Respiratory System

202 David G. Brownstein

Etiology. Sendai virus is a parainfluenza 1 virus of the genus Paramyxovirus, family Paramyxoviri­dae. It is a pleomorphic, enveloped virus measur­ing approximately 100-300nm in length. The he­lical nucleocapsid contains a continuous single strand of RNA. The envelope is studded with two types of surface-projecting glycosylated polypep­tides. The larger projection has hemagglutination and neuraminidase activity; the smaller projec­tion has hemolysin and cell fusion activity. The vi­rus rapidly inactivates at temperatures between 20° and 37°C (Chanock et al. 1963).

Frequency. Sendai virus is ubiquitous in colonies of mice and rats. A survey of various institutional and commercial colonies, published in 1978, indi­cated that 66% of mouse colonies and 63% of rat colonies had experienced infections (Parker et al. 1978). Attack rates usually exceed 50% (Parker et al. 1964, 1978).

Comparison with Other Species

Parainfluenza viruses naturally infect humans, other primates, dogs, cattle, sheep, and birds in addition to rodents. All mammalian parainfluen­za viruses replicate and cause disease primarily in the respiratory tract. They have a tropism for the epithelium of the conducting airways. Besides Sendai virus, histopathology is well described for parainfluenza 3 virus infection in calves (Dawson et al. 1965; Omar et al. 1966; Tsai and Thomson 1975). These lesions are similar to those caused by Sendai virus in rodents. Intranuclear inclusions are more prevalent and intracytoplasmic inclu­sions are more distinct in parainfluenza 3 virus in­fections.

References

Appel LH, Kovatch RM, Reddecliff 1M, Gerone PI (1971) Pathogenesis of Sendai virus infection in mice. Am 1 Vet Res 32: 1835-1841

Bhatt PN, 10nas AM (1974) An epizootic of Sendai infec­tion with mortality in a barrier-maintained mouse col­ony. Am 1 Epidemiol100: 222-229

Brownstein DG, Smith AL, 10hnson EA (1981) Sendai vi­rus infection in genetically resistant and susceptible mice. Am J Patholl05: 156-163

BureklD, ZurcherC, van Nunen MCl, HollanderCF (1977) A naturally occurring epizootic caused by Sendai virus in breeding and aging rodent colonies. II. Infection in the rat. Lab Anim Sci 27: 963-971

Chanock RN, Parrott RH, Johnson KM, Kopikian AZ,

Bell lA (1963) Myxoviruses: parainfluenza. Am 1 Respir Dis 88 (Suppl): 152-166

Choppin PW, Compans RW (1975) Reproduction of par­amyxoviruses. Compr Virol4: 95-178

Darlington RW, Portner A, Kingsbury DW (1970) Sendai virus replication: an ultrastructural comparison of pro­ductive and abortive infections in avian cells. 1 Gen Vi­ro19: 169-177

Dawson PS, Darbyshire IH, Lamont PH (1965) The inocu­lation of calves with parainfluenza 3 virus. Res Vet Sci 6: 108-113

Eustatia 1M, Maase E, van Heiden P, van der Veen 1 (1972) Viral replication in mouse macrophages. Arch Virus­forsch 39: 376-380

Fujiwara K, Takenaka S, Shumiya S (1976) Carrier state of antibody and viruses in a mouse breeding colony per­sistently infected with Sendai and mouse hepatitis vi­ruses. Lab Anim Sci 26: 153-159

Iida T, Tajima M, Murata Y (1973) Transmission of mater­nal antibodies to Sendai virus in mice and its signifi­cance in enzootic infection. 1 Gen Viro118: 247-254

Iwai H, Goto Y, Ueda K (1979) Response of athymic nude mice to Sendai virus. Ipn 1 Exp Med 49: 123-130

lacoby RO, Bhatt PN, 10nas AM (1979) Viral disease. In: Baker HI, Lindsey lR, Weisbroth SH (eds) The labora­tory rat, vol 1, Biology and diseases. Academic, New York, chap 11

lakab Gl (1974) Effect of sequential inoculations of Sen­dai virus and Pasteurella pneumotropica in mice. 1 Am Vet Med Assoc 164: 723-728

lakab Gl, Dick EC (1973) Synergistic effect in viral-bacte­rial infection: combined infection of the murine respira­tory tract with Sendai virus and Pasteurella pneumotropi­ca. Infect Immun 8: 762-768

Mims CA, Murphy FA (1973) Parainfluenza virus Sendai infection in macrophages, ependyma, choroid plexus, vascular endothelium and respiratory tract of mice. Am 1 Pathol 70: 315-328

Omar AR, lennings AR, Betts AO (1966) The experimental disease produced in calves by the J-121 strain of parain­fluenza virus type 3. Res Vet Sci 7: 379-388

Parker JC, Reynolds RK (1968) Natural history of Sendai virus infection in mice. Am J Epidemiol 88: 112-125

Parker lC, RichterCB (1982) Viral diseases of the respira­tory system. In: FosterHL, SmalllD, FoxlG (eds) The mouse in biomedical research, vol 2, Diseases. Academ­ic, New York, chap 8

Parker lC, Tennant, RW, Ward TG, Rowe WP (1964) Enzootic Sendai virus infections in mouse breeder colonies within the United States. Science 146: 936-938

Parker lC, Whiteman MD, Richter CB (1978) Susceptibili­ty of inbred and outbred mouse strains to Sendai virus and prevalence of infection in laboratory rodents. Infect Immun 19: 123-130

Reid WD, lIett KF, Glick 1M, Krishna G (1973) Metabo­lism and binding of aromatic hydrocarbons in the lung. Relationship to experimental bronchiolar necrosis. Am Rev Respir Dis 107: 539-551

RichterCB (1970) Application of infectious agents to the study of lung cancer: studies on the etiology and mor­phogenesis of metaplastic lung lesions in mice. USAEC Symposium Series 21: 365-382

Richter CB (1973) Experimental pathology of Sendai virus infection in mice. 1 Am Vet Med Assoc 163: 1204

Page 213: Respiratory System

Robinson TWE, Cureton RJR, Heath RB (1968) The pa­thogenesis of Sendai virus infection in the mouse lung. J Med Microbiol 1: 89-95

Sawicki L (1961) Influence of age of mice on the recovery from experimental Sendai virus infection. Nature 192: 1258-1259

Sawicki L (1962) Studies on experimental Sendai virus in­fection in laboratory mice. Acta Virol (Praha) 6: 347-351

Stephens RJ, Sloan MF, Evans MJ, Freeman G (1974) Ear­ly response of lung to low levels of ozone. Am J Pathol 74:31-58

Stewart RB, Tucker MJ (1978) Infection of inbred strains of mice with Sendai virus. Can J Microbiol 24: 9-13

Tsai KS, Thomson RG (1975) Bovine parainfluenza type 3 virus infection: ultrastructural aspects of viral pathogen­esis in the bovine respiratory tract. Infect Immun 11: 783-803

van Nunen MCJ, van der Veen J (1967) Experimental in-

Rat Coronavirus Infection, Lung, Rat

David G. Brownstein

Synonym. Parker's rat coronavirus infection.

Gross Appearance

Naturally infected adult rats rarely have grossly observable changes. Experimentally infected 9-10 week old axenic rats develop gross lesions in the lung on postinoculation days 6 and 7, which consist of randomly dispersed red-brown to gray foci, less than 1 mm in diameter (Bhatt and Jaco­by 1977). Although rat coronavirus may cause fa­tal pneumonia in a high percentage of newborn and day-old rats, gross pulmonary lesions have not been described (Parker et al. 1970).

Microscopic Features

Lung changes in young adult rats are mild and short-lived (Bhatt and Jacoby 1977). Bronchus-as­sociated lymphoid tissue is hyperplastic (Fig. 248), some pulmonary veins and venules are cuffed by lymphocytes (Fig.249), and there is patchy inter­stitial pneumonia (Fig. 250). Septa of affected alveoli are thickened by mononuclear cells and neutrophils. Adjacent alveolar spaces contain desquamated pneumocytes, foamy macro phages, lymphocytes, and neutrophils.

Rat Coronavirus Infection, Lung, Rat 203

fection with Sendai virus in mice. Arch Virusforsch 22: 388-397

van der Veen J, Poort Y, Birchfield DJ (1970) Experimental transmission of Sendai virus infection in mice. Arch Vi­rusforsch 31 : 237-246

van der Veen J, Poort Y, Birchfield DJ (1972) Effect of rela­tive humidity on experimental transmission of Sendai virus in mice. Proc Soc Exp Bioi Med 140: 1437-1440

van der Veen J, Poort Y, Birchfield DJ (1974) Study of the possible persistence of Sendai virus in mice. Lab Anim Sci 24: 48-50

Ward JM (1974) Naturally occurring Sendai virus disease of mice. Lab Anim Sci 24: 938-942

WardJM, HouchensDP, CollinsMJ, YoungDM, Rea­gan RL (1976) Naturally-occurring Sendai virus infec­tion of athymic nude mice. Vet Pathol13: 36-46

Zurcher C, Burek JD, van Nunen MCJ, Meihuizen SP (1977) A naturally occurring epizootic caused by Sendai virus in breeding and aging rodent colonies. I. Infection in the mouse. Lab Anim Sci 27: 955-962

Transient rhinotracheitis also occurs and may lead to segmental erosion of the respiratory epi­thelium covering nasal turbinates. The lamina propria is edematous and infiltrated with lympho­cytes and neutrophils. Some nasal respiratory sur­faces are covered with exudate consisting of mu­cus, neutrophils, desquamated epithelium, and detritus. Tracheal epithelium is rarely eroded but large numbers of trans epithelial neutrophils may be present. The lamina propria is mildly edema­tous, congested, and infiltrated with lymphocytes and neutrophils. Lesions of salivary glands are uncommon but may help distinguish rat coronavirus infections from other rat respiratory infections. Mild paroti­tis and submaxillary sialoadenitis have been re­ported and are identical to, but less severe than, those caused by sialodacryoadenitis virus (Bhatt and Jacoby 1977). There is necrosis of salivary ducts with periductular and interstitial inflamma­tory edema. Lesions apparently do not occur in lacrimal glands. Infected neonatal rats develop diffuse interstitial pneumonia, focal atelectasis, and compensatory emphysema (Parker et al. 1970).

Page 214: Respiratory System

204 David G. Brownstein

Page 215: Respiratory System

Ultrastructure

Infected epithelial cells have focally dilated cis­ternae of endoplasmic reticulum and cytoplasmic vacuoles which contain spherical dense cores, 60-70 nm, in diameter, surrounded by an enve­lope 80-120 nm in diameter (Parker et al. 1970; Jonas et al. 1969). The characteristic corona, seen in negatively stained preparations, is not seen by transmission ultramicroscopy. Morphologically, rat coronavirus is indistinguishable from sialo­dacryoadenitis virus.

Differential Diagnosis

Respiratory tract lesions must be differentiated from those caused by sialodacryoadenitis virus, Sendai virus, pneumonia virus, Mycoplasma pul­monis, and pathogenic bacteria. Lesions in sali­vary glands, when present, are generally milder than those caused by sialodacryoadenitis virus.

Biologic Features

Natural History. Rat coronavirus causes acute limited infections of the respiratory tract. The ep­izootiologic characteristic of this infection has not been reported. No evidence for a carrier state has been reported and host range studies have been limited to the rat. The virus is highly infectious and is presumably transmitted by aerosol or direct contact. The infection is subclinical in rats shortly following weaning. Infected neonatal rats may die with severe respiratory distress.

PathogenesiS. Rat coronavirus is epitheliotropic and replicates at all levels of the respiratory tract during the 1st week of infection. The highest titers

<l Fig.248 (Above). Rat coronavirus infection, lung. Bronchi­ole with mildly hyperplastic lymphoid nodules following experimental infection. Hand E, x 59 (reduced by 15%)

Fig. 249 (Middle). Rat coronavirus infection, lung. Periven­ular lymphoid cells in an experimentally infected rat. H and E, x 536 (reduced by 15%)

Fig.250 (Below). Inerstitial pneumonia following experi­mental infection with rat coronavirus. Alveoli contain foamy macrophages and lymphoid cells. Septa are infil­trated with mononuclear cells. Hand E, x 536 (reduced by 15%)

Rat Coronavirus Infection, Lung, Rat 205

are reached in the nasal cavity and trachea. lim­ited replication occurs in salivary tissues. Neu­tralizing antibodies to rat coronavirus and sialo­dacryoadenitis virus are detectable on day 6 or 7. Complement-fixing antibodies appear later and cross-react with sialodacryoadenitis and mouse hepatitis viral antigens (Bhatt and Jacoby 1977; Parker et al. 1970; Jacoby et al. 1979).

Etiology. Rat coronavirus is a typical member of the Coronaviridae: a pleomorphic, enveloped RNA virus with plump, pedunculated surface projections (corona). It measures 76-98 nm in di­ameter in negatively stained preparations with 12-25 nm surface projections (Parker et al. 1970). Virions are formed in cytoplasmic vesicles and cisternae of endoplasmic reticulum. The virus is closely related antigenically to sialodacryoadeni­tis virus (Bhatt et al. 1972).

Frequency. The frequency of rat coronavirus in­fection within rat colonies is difficult to ascertain because of the close relationship of this virus to sialodacryoadenitis virus. Serological evidence of infection with coronaviruses is common in com­mercial and institutional rat colonies. In one sur­vey of 4 germ-free, 5 specific-pathogen-free (SPF), and 11 conventional rat colonies, 3 of the SPF and 11 of the conventional colonies were infected while the germ-free colonies were not (Parker et al. 1970). In another report, all retired breeders from six vendors were positive (Jacoby et al. 1979).

Comparison with Other Species

Coronaviruses are ubiquitous in humans, ani­mals, and birds (Bohl 1981). They cause enteritis in swine, cattle, dogs, mice, turkeys, and humans; encephalomyelitis in swine and mice; systemic in­fections in cats and mice; sialodacryoadenitis in rats; and respiratory infections in chickens, rats, and humans. Avian infectious bronchitis virus in­fects the trachea and lungs of chickens, causing respiratory distress, especially in young chicks. The virus also replicates in kidneys, bursa, and oviducts, producing inflammatory disease at these sites. Human respiratory coronaviruses are apparently restricted to the upper respiratory tract, causing rhinotracheitis and pharyngitis.

Page 216: Respiratory System

206 David G. Brownstein

References

Bhatt PN, Jacoby RO (1977) Experimental infection of adult axenic rats with Parker's rat coronavirus. Arch Vi­rol 54: 345-352

Bhatt PN, Percy DH, Jonas AM (1972) Characterization of the virus of sialodacryoadenitis of rats: a member of the coronavirus group. J Infect Dis 126: 123-130

Bohl EH (1981) Coronaviruses: diagnosis of infections. In: Kurstak E, Kurstak C (eds) Comparative diagnosis of vi­ral diseases, vol 4. Academic, New York, chap 7

Jacoby RO, Bhatt PN, Jonas AM (1979) Viral diseases. In: Baker HJ, Lindsey JR, Weisbroth SH (eds) The labora­tory rat, vol 1. Academic, New York, chap 11

Jonas AM, CraftJ, Black CL, Bhatt PN, Hilding D (1969) Sialodacryoadenitis in the rat. A light and electron mi­croscopic study. Arch Pathol88: 613-622

Parker JC, Cross SS, Rowe WP (1970) Rat coronavirus (RCV): a prevalent, naturally occurring pneumotropic virus of rats. Arch Virusforsch 31: 293-302

Pneumonia Virus of Mice Infection, Lung, Mouse and Rat

David G. Brownstein

Synonyms. Mouse pneumonia virus; pneumonia VlruS.

Gross Appearance

Mice. Gross lung changes are not seen in mice naturally infected with pneumonia virus of mice (PVM). Pulmonary consolidation has been pro­duced experimentally using tissue-culture-adapt­ed virus (Harter and Choppin 1967; Tennant et al. 1965) or serially blind-passed PVM-infected lung tissue (Horsfall and Hahn 1940; Curnen and Horsfall 1947). Initially, consolidation is hilar in distribution with subsequent radiation along bronchioles. Consolidated foci are dark red and exude sanguinous fluid when cut. Later these foci become gray.

Rats. Naturally infected adult rats usually have no gross lung changes but may have focal or mul­tifocal plum-colored to gray foci less than 2 mm in diameter. These may occur in any lobe.

Microscopic Features

Mice. Histopathologic lung changes are rare in naturally infected mice. Lesions produced experi­mentally with tissue-culture-adapted (Carthew and Sparrow 1980a; Vogtsberger et al. 1982) or serially blind-passed infected lung tissue (Horsfall and Hahn 1940) include both airway and paren­chymal changes. PVM adapted to BHK-21 cells and inoculated at high doses (104-105 TCID50) in-

tranasally causes a mild erosive bronchiolitis and interstitial pneumonia. Bronchiolar epithelium first develops granular eosinophilic cytoplasm followed by lifting from the basal lamina. Des­quamated epithelium and neutrophils may plug affected airways. Alveolar changes generally lag behind those in the airways. Alveolar septa are edematous, congested, and infiltrated with neu­trophils and macrophages. There is some necrosis within alveolar walls. By the time bronchiolar epi­thelium has regenerated and returned to a normal appearance, alveolar septa are thickened by dense infiltrates oflymphocytes, macrophages, and scat­tered neutrophils. Alveolar spaces may contain an exudate with a similar inflammatory cell compo­sition. Inflammatory changes in the parenchyma peak near the end of the 2nd week of experimen­tal infection and are usually resolved by the end of the 3rd week. Lower doses « 103 TCID50) of tissue-culture­adapted PVM cause vascular-oriented inflamma­tion and interstitial pneumonia (Vogtsberger et al. 1982). The vascular component has been de­scribed as an acute vasculitis with infiltrates of neutrophils and lymphocytes followed by a mild nonsuppurative vasculitis, which persists to the end of the 3rd week of infection. The interstitial pneumonia is acute, but its duration and histolog­ical features have not been reported. Mild rhinitis is a constant feature in mice infected intranasally with tissue-culture-adapted PVM, even at doses that fail to produce pulmonary his­topathology. The nasal mucosa is edematous with multi focal erosions. Neutrophils and lymphoid cells infiltrate the lamina propria. A sparse ex-

Page 217: Respiratory System

Pneumonia Virus of Mice Infection, Lung, Mouse and Rat 207

Fig.251 (Above). Perivascular infiltrates and interstitial pneumonia in a rat naturally infected with PVM. Hand E, x 64 (reduced by 15%)

udate rich in neutrophils and desquamated epi­thelium is occasionally present.

Rats. Histopathologic changes in lung have been observed in naturally and experimentally infected rats. Naturally infected weanling Lewis rats devel­op hyperplasia of bronchus-associated lymphoid

Fig.252 (Below). Perivascular plasma cell infiltrates in a PVM-infected rat. Hand E, x 217 (reduced by 15%)

tissue (BAL T), perivascular mononuclear cell in­filtrates, and multifocal interstitial pneumonia (Fig. 251). Prominent germinal centers form with­in the expanded BALT. The overlying airway epi­thelium is intact but bulges into the lumen. Trans­epithelial lymphocytes are increased near these reactive lymphoid nodules. Pulmonary venules,

Page 218: Respiratory System

208 David G. Brownstein

Fig. 253. Interstitial pneumonia in a PVM-infected rat. Alveolar septa are congested, edematous, and infiltrated with mixed mononuclear cells. Alveolar lining cells are hy-

small veins, and aterioles contain dense, usually symmetrical, adventitial infiltrates of plasma cells, reactive lymphoid cells, and macrophages (Fig.252). The endothelium of these vessels may be hypertrophied and leukocyte pavementing may be prominent. Perivascular infiltrates often occur in areas of interstitial inflammation. Alveo­lar septa are congested, edematous, and infiltrat­ed with lymphocytes, plasma cells, and macro­phages. Alveolar lining cells are swollen or hy­pertrophied. Alveolar spaces contain variable numbers of foamy macrophages, desquamated pneumocytes, lymphocytes, and neutrophils (Fig. 253). Multifocal nonsuppurative vasculitis and acute interstitial pneumonia have been described in experimentally infected Fischer 344 rats (Vogts­berger et al. 1982).

Ultrastructure

The fine structure of PVM infection has only been described in cell cultures (Compans et al. 1967; Berthiaume et al. 1974). Nucleocapsid assembly occurs in the cytosol. Pleomorphic inclusions, which may be seen by light microscopy in the vi­cinity of the nucleus, are composed of electron­dense dots or threads approximately 12 nm in di-

pertrophied. Alveolar spaces contain large foamy mono­nuclear cells - probably desquamated pneumocytes and macrophages. Hand E, x 536 (reduced by 15%)

ameter. Virus maturation occurs at the plasma membrane, where budding particles have round or filamentous profiles about 80-120 nm in di­ameter with filamentous forms predominating. The viral envelope contains surface projections approximately 12 nm in length. Within the en­velope are four to eight electron-dense dots or strands identical to those seen in the cytosol. Strands are usually coiled in filamentous forms. Terminal swellings, 150-300 nm in diameter, are usually present on filamentous forms. In negatively stained preparations, filamentous forms predominate with lengths up to 3 !-Lm and diameters of approximately 100 nm.

Differential Diagnosis

Mice. Naturally occurring PVM apparently rarely causes pneumonia. Until there is evidence to the contrary, PVM should be considered an unlikely cause of inflammatory lung disease in mice under natural conditions, but should be regarded as a cause of mild erosive rhinitis.

Rats. The pulmonary changes in PVM infection must be distinguished from those caused by Sen­dai virus, rat coronaviruses, Mycoplasma pulmo­nis. and pathogenic bacteria. The lesions caused

Page 219: Respiratory System

Pneumonia Virus of Mice Infection, Lung, Mouse and Rat 209

by PVM most closely resemble those caused by pneumotropic strains of rat coronaviruses (Park­er's rat coronavirus). Generally, the latter causes milder lesions than PVM.

Biologic Features

Natural History. Pneumonia virus of mice causes acute limited infections in immunocompetent ro­dents. Virus peristence (over 20 days) has been re­ported in germ-free athymic (nu/nu) mice (Car­thew and Sparrow 1980b). Enzootic and epizootic forms exist and both are asymptomatic in mouse and rat colonies. Transmissibility in mouse colonies is low; infec­tions may therefore be focal (Tennant et al. 1966). Attack rates in 59% of infected mouse colonies are reported to be 25% or less. This is the lowest attack rate for any indigenous murine virus (Ten­nant et al. 1966). Different age groups may harbor the infection, depending on management prac­tices. In one colony, antibody was first detected in 8-week-old mice, while in a second colony, mice did not seroconvert until the age of 7 months (Parker et al. 1966). Attack rates are apparently higher in rat colonies. We frequently find a 100% prevalence of serum antibodies (hemagglutination inhibition test) in weanling rats from enzootically infected colo­meso Clinical signs have not been reported for natu­rally infected mice or rats. Depression, anorexia, weight loss, ruffled fur, hunched posture, and la­bored respiration have been reported in experi­mentally infected mice (Horsfall and Hahn 1940).

Pathogenesis. Viral replication is restricted to the epithelium of the respiratory tract (Carthew and Sparrow 1980a, b). Infectious virus can be detect­ed up to 10 days after exposure (A. L. Smith and V. A. Carrano personal communication). Viral an­tigens have not been detected in lung sections be­yond day 7 in mice (Carthew and Sparrow 1980a). Virus is most consistently isolated from nasal washes in experimentally or naturally in­fected rodents (A. L. Smith and V. A. Carrano, per­sonal communication). Seroconversion (hemagglutination inhibition [HAl], complement fixation [CF]) usually occurs 9 or 10 days after exposure. CF antibody titers be­gin to decline during the 3rd week of infection, while HAl antibody titers remain elevated for at least 4 months (Tennant et al. 1966).

Etiology. Pneumonia virus of mice is a Pneumovi­rus of the family Paramyxoviridae. It shares this genus with respiratory syncytial virus, which has structural and biologic but not antigenic similari­ties (Joncas et al. 1969; Berthiaume et al. 1974). PVM is a predominantly filamentous enveloped virus containing a single-stranded RNA genome. The hem agglutinins probably occur within the fringe of envelope projections. The virus is labile in the environment and rapidly inactivates at room temperature.

Frequency. Pneumonia virus of mice is prevalent in mouse and rat colonies throughout the world. In a recent survey, 63% of mouse and 68% of rat colonies from institutional and commercial sources were infected (Parker and Richter 1982).

Comparison with Other Species

Pneumonia virus of mice and respiratory syncy­tial virus (RSV) are the sole representatives of the genus Pneumovirus. PVM infects rodents natural­ly; RSV infects children, cattle, and sheep, and experimentally infects ferrets and cotton rats. Both of these viruses have potentially broad respi­ratory epitheliotropism that is differentially ex­pressed depending on the host, host age, and in-, fecting dose of virus. Experimentally, PVM causes rhinitis, bronchiolitis, or interstitial pneumonia in mice. Naturally occuring PVM infection in rats causes interstitial pneumonia. RSV in humans and cattle causes rhinitis, bronchiolitis, or intersti­tial pneumonia (Aherne et al. 1970; Mohanty et al. 1975). Experimentally, RSV produces rhinitis and interstitial pneumonia in infant ferrets (Prince and Porter 1976) and bronchiolitis in cot­ton rats (Prince et al. 1978).

References

Aherne W, Bird T, Court SD, Gardner PS, McQuillin J (1970) Pathological changes in virus infections of the lower respiratory tract in children. J Clin Pathol 23: 7-18

Berthiaume L, Joncas J, Pavilanis V (1974) Comparative structure, morphogenesis and biological characteristics of the respiratory syncytial (RS) virus and the pneu­monia virus of mice (PVM). Arch Virusforsch 45: 39-51

Carthew P, Sparrow S (1980a) A comparison in germ-free mice of the pathogenesis of Sendai virus and mouse pneumonia virus infections. J Pathol130: 153-158

Carthew P, Sparrow S (1980b) Persistence of pneumonia

Page 220: Respiratory System

210 David. G. Brownstein

virus of mice and Sendai virus in germ-free (nu/nu) mice. Br J Pathol61: 172-175

Compans RW, Harter DH, Choppin PW (1967) Studies on pneumonia virus of mice (PVM) in cell culture. II. Struc­ture and morphogenesis of the virus particle. J Exp Med 126:267-276

Cumen EC, Horsfall FL Jr (1947) Properties of pneumonia virus of mice (PVM) in relation to its state. J Exp Med 85:39-53

Harter DH, Choppin PW (1967) Studies on pneumonia vi­rus of mice (PVM) in cell culture. I. Replication in baby hamster kidney cells and properties of the virus. J Exp Med 126: 251-266

Horsfall FL, Hahn RG (1940) A latent virus in normal mice capable of producing pneumonia in its natural host. J Exp Med 71: 391-408

Joncas J, Berthiaume L, Pavilanis V (1969) The structure of the respiratory syncytial virus. Virology 38: 493-496

Mohanty SB, Ingling AL, Lillie MG (1975) Experimentally induced respiratory syncytial viral infection in calves. Am J Vet Res 36: 417-419

Parker JC, Richter CB (1982) Viral diseases of the respira-

tory system. In: FosterHL, SmallJD, FoxJG (eds) The mouse in biomedical research, vol 2, Diseases. Academ­ic, New York, chap 8

ParkerJC, Tennant RW, Ward TG (1966) Prevalence of vi­ruses in mouse colonies. Natl Cancer Inst Monogr 20: 25-36

Prince GA, Porter DD (1976) The pathogenesis of respira­tory syncytial virus infection in infant ferrets. Am J Pathol82: 339-352

Prince GA, Jenson AB, Horswood RL, Camargo E, Cha­nock RM (1978) The pathogenesis of respiratory syncy­tial virus infection in cotton rats. Am J Pathol 93: 771-791

Tennant RW, Parker JC, Ward TG (1965) Virus studies with germ-free mice. II. Comparative responses of germ-free mice to virus infection. JNCI 34: 381-387

Tennant RW, Parker JC, Ward TG (1966) Respiratory virus infections of mice. Natl Cancer Inst Monogr 20: 93-104

Vogtsberger LM, Stromberg PC, Rice JM (1982) Histologi­cal and serological response of B6C3F1 mice and F344 rats to experimental pneumonia virus of mice infection. Lab Anim Sci 32: 419 (abstract)

Sialodacryoadenitis Virus Infection, Lung, Mouse

David G. Brownstein

Synonyms. Rat submaxillary gland virus infec­tion; SDAV infection.

Gross Appearance

Weanling mice experimentally infected by the in­tranasal route develop red-brown foci distributed uniformly over all lobes of the lung (Bhatt et al. 1977).

Microscopic Features

The typical pulmonary lesion is multifocal inter­stitial pneumonia. This lesion is acute, with a peak intensity on postexposure day 6 and resolution beginning on days 8-10. Inflammatory foci are usually oriented around terminal bronchioles (Fig.254) and frequently radiate to the pleura. In some cases, however, orientation around terminal airways is not obvious. In these cases, inflamma­tion is randomly distributed and usually less cir­cumscribed than peribronchiolar inflammatory foci.

Initially, septa of affected alveoli are edematous and congested with reactive or degenerative changes in lining epithelium. Reactive cells are hypertrophied with large vesicular nuclei; degen­erating cells are pyknotic and sloughing. Macro­phages and, to a lesser degree, lymphocytes accu­mulate in the interstitium and alveolar spaces (Fig. 255). The adventitia of blood vessels adja-

Fig.254 (Above). Sialodacryoadenitis viral pneumonia in l> an experimentally infected weanling mouse. Inflammation is oriented around terminal bronchioles. Hand E, x 54 (reduced by 15%)

Fig. 255 (Middle). Sialodacryoadenitis viral infection. Early pneumonic changes in an experimentally infected mouse. Alveolar septa are congested and edematous with in­creased cellularity due to lymphoreticular infiltrates. At this magnification occasional swollen lining cells can be identified. Hand E, x 195 (reduced by 15%)

Fig.256 (Below). Sialodacryoadenitis viral infection. Peak pneumonic changes in an experimentally infected mouse. Alveolar septa are expanded by lymphoreticular cells and a modest mononuclear cell exudate is in the alveoli. Hand E, x 195 (reduced by 15%)

Page 221: Respiratory System

Sialodacryoadenitis Virus Infection, Lung, Mouse 211

Page 222: Respiratory System

212 David. G. Brownstein

cent to inflammatory foci contains loose eccentric aggregates of mixed mononuclear cells. Leuko­cyte pavementing is usually seen along the intima of these vessels. Terminal airways, around which inflammatory foci are oriented, rarely have de­tectable change in lining epithelium. During the period of peak inflammation, alveolar septa are markedly thickened by lymphoreticular cells (Fig. 256). Foamy macrophages, frequently in cohesive clusters, are numerous in alveolar spaces along with desquamated pneumocytes, some lymphocytes, and necrotic debris. Partial atelectasis may be present in severely inflamed foci. Resolution begins with a decline in the cellularity and thickness of alveolar septa. Foamy macro­phages continue for a while to be numerous in alveolar spaces. Transient lymphoid nodules and diffuse lymphoplasmacytic infiltrates develop in the adventitia of nearby blood vessels and bron­chioles. The duration of these changes is not known.

Ultrastructure

Ultramicroscopic changes have not been report­ed for sialodacryoadenitis virus infection in mice. The appearance of this virus in trans­mission electron micrographs of infected rat submaxillary glands has been reported (Jonas et al. 1969). Within infected cells, cisternae of endoplasmic reticulum and cytoplasmic vesicles contain dense or hollow spherical cores, 60-70 nm in diameter, surrounded by an enve­lope 40-120 nm in diameter. The corona seen in negatively stained preparations is not seen by transmission ultramicroscopy.

Differential Diagnosis

Sialodacryoadenitis viral pneumonia in mice must be distinguished from pneumonia caused by Sendai virus, pneumonia virus, K virus, mouse coronavirus, Mycoplasma pulmonis, and patho­genic bacteria such as Corynebacterium kutscheri. The alveolar lesion seen in SDAV infection re­sembles the alveolar component of Sendai virus infection but is less exudative, and is not accom­panied by inflammatory airway changes. Both SDAV and certain strains of mouse coronavirus (mouse hepatitis virus) cause inflammatory lung lesions and seroconversion to mouse hepatitis vi­rus in mouse colonies (Bhatt et al. 1977). Because

mice are apparently not natural hosts of sialoda­cryoadenitis virus, contact with infected rats is probably required.

Biologic Features

Natural History. This virus has not been proved to infect mice under natural conditions. Some epidemiologic evidence indicates that sialodac­ryoadenitis virus may account for unexpected or unexplained appearance of antibodies to mouse hepatitis virus in mouse colonies (Jacoby et al. 1979). In rats SDAV is highly contagious by aerosol, contact, and fomites. Whether mouse to mouse transmission is equally efficient has not been determined. Mice are asymptomatic dur­ing experimental SDAV infections (Bhatt et al. 1977).

Pathogenesis. This virus causes acute limited in­fections in experimentally inoculated mice. The virus is epitheliotropic with replication limited to the respiratory tract. It replicates at all levels of the respiratory tract but produces disease pri­marily in the lungs, where the highest viral titers are achieved. The tissue tropism of SDAV in mice differs from that in infected rats. In rats, virus replication and disease occur primarily in exocrine tissues of the head and epithelium of the upper respiratory tract. In mice, the princi­pal targets are alveolar lining cells, with poor re­plication of virus in epithelium of the upper re­spiratory tract. Virus titers peak in the lungs on postexposure day 2 and are not detectable by day 8 (Bhatt et al. 1977).

Etiology. Sialodacryoadenitis virus, one of the Coronaviridae, is a pleomorphic, enveloped RNA virus with plump, pedunculated surface projec­tions (corona). It is approximately 114nm in di­ameter (Jonas et al. 1969). The virus replicates in­tracytoplasmically and virions are formed in cytoplasmic vesicles and endoplasmic reticulum (Jacoby et al. 1979). The virus is closely related antigenically to Parker's rat coronavirus (Bhatt et al. 1972).

Frequency. Coronavirus infections are common in commercial and institutional mouse colonies. Most of these infections are due to mouse coro­naviruses (mouse hepatitis virus), but the possibil­ity that some may result from SDA V cannot be ruled out; some mice are clearly susceptible to experimental infection with the virus.

Page 223: Respiratory System

Comparison with Other Species

Coronaviruses are widespread in humans, ani­mals, and birds. They produce enteritis, encepha­lomyelitis, sialodacryoadenitis, and systemic in­fections in addition to infections of the respiratory tract. Generally, respiratory coronaviruses, which infect chickens, rats, and humans, produce dis­ease in the upper respiratory tract. There is evi­dence in humans, however, that coronaviruses are important causes of viral pneumonia (Mcintosh et al. 1974).

Murine Respiratory Mycoplasmosis, Lung, Rat 213

References

Bhatt PH, Percy DH, Jonas AM (1972) Characterization of the virus of sialodacryoadenitis of rats: a member of the coronavirus group. J Infect Dis 126: 123-130

Bhatt PN, Jacoby RO, Jonas AM (1977) Respiratory infec­tion in mice with sialodacryoadenitis virus, a coronavi­rus of rats. Infect Immun 18: 823-827

Jacoby RO, Bhatt PN, Jonas AM (1979) Viral diseases. In: Baker HJ, Lindsey JR, Weisbroth SH (eds) The labora­tory rat, vol 1. Academic, New York, chap 11

Jonas AM, CraftJ, Black CL, Bhatt PN, Hilding D (1969) Sialodacryoadenitis in the rat. A light and electron mi­croscopic study. Arch Pathol88: 613-622

McIntosh K, Chao RK, Krause HE, Wasil R, Mocega HE, Mufson MA (1974) Coronavirus infection in acute lower respiratory tract disease of infants. J Infect Dis 130: 502-507

Murine Respiratory Mycoplasmosis, Lung, Rat

Trenton R. Schoeb and 1. Russell Lindsay

Synonyms. Murine chronic respiratory disease; chronic murine pneumonia; enzootic bronchiec­tasis.

Gross Appearance

The lungs are externally normal in the majority of infected rats, gross lesions being poorly correlated with clinical signs and microscopic changes. The causative organism, Mycoplasma pulmonis, pref­erentially affects the nasal passages and middle ears, and the incidence of lesions decreases from the nose distally. The bronchi are the most com­monly affected parts of the lungs. Purple, de­pressed areas of atelectasis may occur in lungs in which exudate obstructs airways. A few rats with advanced disease have gray- or yellow-purple consolidated areas of pneumonia. Yellow, slightly elevated foci representing bronchioles dilated with purulent exudate can affect entire lobes, im­parting a cobblestone appearance (Figs.257 and 258). Frank abscesses also are seen in a few cases. The bronchial and paratracheallymph nodes may be enlarged to three or four times normal size.

Fig. 257. Murine respiratory mycoplasmosis, rat. Severe diffuse bronchiolectasis, atelectasis, and pneumonia in the left lung. x 2

Page 224: Respiratory System

214 Trenton R. Schoeb and 1. Russell Lindsay

Microscopic Features

The lesions of murine respiratory mycoplasmosis are so common that many pathologists lack famil­iarity with the histology of normal rat lungs (Figs.259 and 260), which differs slightly from that of many other species. First, pathogen-free and even germfree laboratory rats have small amounts of bronchus-associated lymphoid tissue, especially at the acute angles of bronchial bifurca­tions, and there are also lymphoid aggregates be­tween the bronchi and adjacent blood vessels. Second, the trachea, bronchi, and bronchioles are lined by cuboidal to low columnar epithelium, rather than the tall columnar, pseudostratified type. Lesions in the lung usually occur mainly in the major airways and are characterized by neutro-

Fig.258. Murine respiratory mycoplasmosis, rat. Severely affected left lung with bronchiectasis, bronchiolectasis, atelectasis, pneumonia, and greatly increased bronchial lymphoid tissue. x 8

philic exudate, epithelial hyperplasia, and hyper­trophy in varying degrees, and an increase in peri­bronchial lymphoid tissue (Figs. 261-263). The distribution and severity of these changes within the lungs, and even within an individual lobe, are quite variable. Bronchiectasis and bronchiolecta­sis often result as airways become distended with purulent exudate. After weeks or months, the epi­thelium of these distended airways becomes flat­tened ("squamoid") or even nonkeratinizing strat­ified squamous. Less commonly, the epithelium is destroyed completely, and lost in the resulting ab­scess. The epithelium of alveoli around these se­verely affected airways may become cuboidal, im­parting a glandular appearance (Fig. 263). Vari­ably distributed alveolar exudation of neutrophils and macrophages is a feature of advanced disease in which infection has spread centrifugally be­yond the bronchioles.

..

Fig. 259. Normal lungs of a rat with small amounts oflym­phoid tissue predominantly at major bronchial bifurca­tions. x 4 (reduced by 10%)

Page 225: Respiratory System

Ultrastructure

Mycoplasma pulmonis parasitizes the surface of respiratory epithelial cells. Heavy infections in­duce degenerative changes, such as loss of cilia and cytoplasmic vacuolation. Some cells also un­dergo more severe changes, indicating irreversible damage (necrosis), but it is not established wheth­er such damage is due to the organism, to asso­ciated inflammatory processes, or both.

Differential Diagnosis

In naturally occurring respiratory disease in rats, M.pulmonis is almost universally present. One or more other bacteria also may be found, such as Streptococcus pneumoniae, Corynebacterium

Fig. 260. Normal bronchial lymphoid tissue of rat with flat epithelium and mostly small lymphocytes. Hand E, x 450

Murine Respiratory Mycoplasmosis, Lung, Rat 215

kutscheri, Bordetella bronchispetica, Pasteurella pneumotropica, Streptobacillus moniliformis, Pseu­domonas aeruginosa, Klebsiella pneumoniae, and an unidentified argyrophilic bacillus which also parasitizes the surface of respiratory epithelial cells (MacKenzie et al. 1981; van Zwieten et al. 1980). Most of these organisms are probably only oppor­tunistic pathogens but S. pneumoniae and C. kutscheri are considered to be primary pathog­ens (Weisbroth 1979). S.pneumoniae causes fi­brinopurulent bronchopneumonia, pleuritis, and pericarditis, and C. kutscheri induces multifocal suppurative pneumonia. Both organisms can be demonstrated in sections by tissue gram stains. These diseases are easily differentiated from mu­rine respiratory mycoplasmosis, but because af­fected rats commonly have concurrent mycoplas-

Fig. 261. Mildly affected lungs of a rat with mild to moder­ate increase in bronchial lymphoid tissue and early bron­chiolitis in the right cranial lobe and cranial part of the left lung. x 4

Page 226: Respiratory System

216 Trenton R. Schoeb and J. Russell Lindsay

mal disease, cultural, morphological, and serolog­ic evidence of respiratory mycoplasmosis should still be sought. Many rats with respiratory mycoplasmosis also have concurrent Sendai virus, sialodacryoadenitis virus, or rat coronavirus infection. Sendai virus in­fection in adult rats is usually subclinical. Lesions are similar to those in mice and are characterized by necrotizing bronchiolitis (Jacoby et al. 1979). Sialodacryoadenitis and rat coronavirus have been reported to cause multifocal interstitial pneumonia in natural or experimental infections, but these agents to no appear to be important re­spiratory pathogens for rats. None of these le­sions, if found, should be difficult to distinguish from those of mycoplasmosis. An accurate diagnosis requires diligent efforts to

Fig.262. Bronchus in murine respiratory mycoplasmosis with neutrophilic exudate, mild hyperplasia of respiratory epithelium, intraepitheliallymphocytes, and large lympho­cytes and plasmacytoid cells in the lamina propria. Hand E, x 450

determine which viruses and bacteria are present, using several methods including microscopic ex­amination of tissues, bacterial and mycoplasmal culturing, and serologic testing. One must criti­cally evaluate and correlate all results to make di­agnoses appropriate to each animal or colony. Failure to isolate M. pulmonis is not evidence of its absence, inasmuch as there are many difficulties in isolating this organism by ordinary methods. For example, certain tissue substances and even medium components can be inhibitory (Del Giu­dice et al. 1980; Kaklamanis et al. 1971; Mardh and Taylor-Robinson 1973; Tauraso 1967). Cul­turing several sites in the respiratory tract is ad­vantageous, as is combining culture with other di­agnostic methods (Davidson et al. 1981), such as enzyme-linked immunsorbent assay (Horowitz

• • •

"PI • • . ,:,'."- .~ ~ I . •• ., ' • . . ;

.to. ~ •

Fig.263. Bronchiole in severe murine respiratory myco­plasmosis with neutrophilic exudate, severe epithelial dis­tortion and ciliary loss, lymphoid accumulation, and peri­bronchiolar glandular structures. Hand E, x 450

Page 227: Respiratory System

and Cassell 1978), which is now available com­mercially. Although microbial agents present si­multaneously with M. pulmonis probably modify the expression of natural respiratory disease in rats, no other agent has been demonstrated to pro­duce lesions resembling those of mycoplasmosis in rats that are clearly free of other pathogens. Therefore, M. pulmonis should be considered the primary pathogen in rats having lesions consistent with those of experimental murine respiratory mycoplasmosis.

Biologic Features

The biologic features are discussed further on page 80. The roles of the various specific and nonspecific host defense mechanisms in resistance of rats to mycoplasmosis are unclear. Systemic antibody and cellular responses occur, but the cellular re­sponse appears more important in rats inasmuch as resistance can be conferred by transfer of cells but not serum (Cassell et al. 1973). Vaccination studies have shown that resistance to disease can be induced by local or systemic vaccination, and have provided circumstantial evidence for the ex­istence of both local and systemic responses (Cas­sell and Davis 1978). A vigorous local response would seem to be indicated by the charateristic lymphoid accumulations; however, much of this could be due to nonspecific mitogenic activity of M. pulmonis (N aot et al. 1979). It remains to be de­termined whether the lymphoid accumulation is due to infiltration, local proliferation, or both. In vivo studies have indicated that alveolar macro­phages may be important in resistance to alveolar invasion by M.pulmonis (Cassell et al. 1973). However, in vitro experiments have demonstrated only inhibition of multiplication of M.pulmonis by alveolar macrophages rather than rapid killing of the organism (Davis et al. 1980). Like other respiratory mycoplasmas, M. pulmonis parasitizes the surface of ciliated epithelial cells. The mechanisms by which it affects these cells are unclear, but possibilities include competition for metabolites or components of the host cells and production of toxic wastes (Cassell et al. 1978). The close association with host cells may contrib­ute to the ability of M.pulmonisto escape elimina­tion by host defenses (Cassell et al. 1978). For ex­ample, it may prevent mucociliary clearance, phagocytosis, or efficient attack by specific im­mune effector mechanisms. However, alteration of lymphocyte responsiveness and consequent

Murine Respiratory Mycoplasmosis, Lung, Rat 217

misdirection or disruption of immune responses by nonspecific mitogenicity (Naot et al. 1979) also seem likely contributors to the organism's vir­ulence, ability to resist elimination, or both (Cas­sell et al. 1979). Mycoplasma pulmonis infection is ubiquitous in conventional rat colonies, and has been identified in "barrier-maintained" colonies in the United States and Great Britain by serologic testing and cultural isolation (Cassell et al. 1981). It also has been found in colonies thought to be germ free (Ganaway et al. 1973).

Comparison with Other Species

Comparative aspects of mycoplasmosis in several species are discussed on page 82.

References

Cassell GH, Davis JK (1978) Protective effect of vaccina­tion against Mycoplasma pulmonis respiratory disease in rats. Infect Immun 21: 69-75

Cassell GH, Lindsey JR, Overcash RG, Baker HJ (1973) Murine mycoplasma respiratory disease. Ann NY Acad Sci 225: 395-412

Cassell GH, Davis JK, Wilborn WH, Wise KS (1978) Pathobiology of mycoplasmas. In: Schlessinger D (ed) Microbiology 1978. American Society for Microbiology, Washington DC, pp 399-403

Cassell GH, Lindsey JR, Baker HJ, Davis JK (1979) Myco­plasmal and rickettsial diseases. In: Baker HJ, Lind­sey JR, Weisbroth SH (eds) The laboratory rat, vol!. Academic, New York, chap 10

Cassell GH, Lindsey JR, Davis JK, Davidson MK, Brown MB, Mayo JG (1981) Detection of natural Myco­plasma pulmonis infection in rats and mice by an enzyme linked immunosorbent assay (ELISA). Lab Anim Sci 31:676-682

Davidson MK, Lindsey JR, Brown MB, Schoeb TR, Cas­sell GH (1981) Comparison of methods for detection of Mycoplasma pulmonis in experimentally and naturally infected rats. J Clin Microbiol14: 646-655

Davis JK, Delozier KM, Asa DK, Minion FC, Cassell GH (1980) Interactions between murine alveolar macro­phages and Mycoplasma pulmonis in vitro. Infect Im­mun 29: 590-599

Del Giudice RA, Gardella RS, Hopps HE (1980) Cultiva­tion of formerly noncultivable strains of Mycoplasma hyorhinis. CUff Microbiol4: 75-80

Ganaway JR, Allen AM, Moore TD, Bohner HJ (1973) Natural infection of germ-free rats with Mycoplasma pulmonis. J Infect Dis 127: 529-537

Horowitz SA, Cassell GH (1978) Detection of antibodies to Mycoplasma pulmonis by an enzyme linked immuno­sorbent assay. Infect Immun 22: 161-170

Jacoby RO, Bhatt PN, Jonas AM (1979) Viral diseases. In: Baker HJ, Lindsey JR, Weisbroth SH (eds) The labora­tory rat, vol 1. Academic, New York, chap 11

Page 228: Respiratory System

218 1. K. Frenkel

Kaklamanis E, Stavropoulos K, Thomas L (1971) The my­coplasmacidal action of homogenates of normal tissues. In: MadoffS (ed) Mycoplasma and the L-forms ofbac­teria. Gordon and Breach, New York, pp27-35

MacKenzie WF, Magill LS, Hulse M (1981) A filamentous bacterium associated with respiratory disease in wild rats. Vet Pathol18: 836-839

Mardh PA, Taylor-Robinson D (1973) New approaches to the isolation of mycoplasmas. Med Mikrobiol Immunol (Beri) 158: 259-266

Naot Y, Merchav S, Ben-David E, Ginsburg H (1979) Mitogenic activity of Mycoplasma pulmonis. I. Stimu-

Pneumocystosis, Lung, Rat

J. K. Frenkel

Synonym. Interstitial plasma cell pneumonia.

Gross Appearance

Heavily infected lungs are focally to diffusely consolidated but never involve an entire lobe. The alveoli are filled with grayish material, between which the pinkish alveolar walls can be seen with a hand lens (Fig. 264). If tissue necrosis or ab­scesses are present, another organism should also

Fig.264. Pneumocytosis, lung of rat. The dorsal aspect of both lungs is diffusely pale pink opaque with only few alveoli containing trapped air (light, refractile areas). Rat

lation of rat Band T lymphocytes. Immunology 36: 399-406

Tauraso NM (1967) Effect of diethylaminoethyl dextran on the growth of mycoplasma in agar. J Bacteriol 93: 1559-1564

van Zwieten MJ, Solleveld HA, Lindsey JR, de Groot FG, Zurcher C, Hollander CF (1980) Respiratory disease in rats associated with a filamentous bacterium: a prelimi­nary report. Lab Anim Sci 30: 215-221

Weisbroth SH (1979) Bacterial and mycotic diseases. In: Baker HJ, Lindsey JR, Weisbroth SH (eds) The labora­tory rat, vol 1. Academic, New York, chap 9

be looked for. In contrast to consolidated lungs, the lungs are not hyperemic and are less volumi­nous. No fibrinous pleuritis occurs unless bacteri­al or fungal pneumonia is present.

Microscopic Features

The alveoli are filled with masses of Pneumocyst­sis organisms, usually with a few accompanying macrophages (Figs. 265 and 266). In hematoxylin-

was treated with 25 mg cortisone acetate twice weekly and with 1 mg amphotericin subcutaneously three times weekly for 75 days (reduced by 30%)

Page 229: Respiratory System

and eosin-stained sections the intraalveolar masses appear regularly vacuolated, comparable to a honeycomb (Fig. 266). With the periodic acid­Schiff technique the honeycombed material is more intensely stained; hematoxylin-stained nu­clei are less distinct (Fig.267). Using the Grocott modification of Gomori's methenamine silver technique, black yeast-like cysts measuring 3-5 11m in diameter are found individually or in groups in some of the alveolar masses (Fig. 268). The number of these cysts is variable: it is less than the number of nuclei staining with hematox­ylin and many alveoli with honeycombed masses contain few or no cysts. The silver technique gen­erally demonstrates only the cyst wall, not the en­closed nuclei (Fig. 269). The cysts are best identi­fied in Giemsa-stained imprints, where they appear as clear, round spaces against a dark-stain­ing protein background with eight dark-staining nuclei. Methyl violet, toluidine blue, and related dyes also stain the cyst walls. Several rapid stain­ing techniques have been described (see, for ex­ample, Macher et al. 1983).

Ultrastructure

Trophozoites are ameboid in shape and measure 1-5 11m with a 20-30 nm pellicle, apparently com­posed of a double layer. Filopodia extend from the surfaces. The nucleus is bordered by a single­or double-layered membrane which is poorly de­fined. The cytoplasm contains mitochondria, ri­bosomes, endoplasmic reticulum, some vacuoles, lipid globules, and granules, some of which stain for glycogen (Campbell 1972). A precyst, limited by a semirigid trilayered membrane, contains one to several masses of nucleoplasm. Cysts measure 3.5-5 11m in diameter and are also limited by a trilayered wall, 50-300 nm in thick­ness. An inner unit type membrane is 5-7 nm thick. A middle granular electrolucent layer usually measures 30-40 nm in diameter, with oc­casional thicker portions. The outer layer is gran­ular, electron dense, and 20-75 nm in thickness. It contains several nuclear masses or separate intra­cystic bodies, mitochondria, glycogen, and vacu­oles. Filopodia may be attached to the surface (Campbell 1972). Intracystic bodies measure 1-2 11m in diameter and are enclosed by a double-layered membrane 20-30 nm thick. Their nuclei measure less than 111m. The nuclear envelope is sometimes continu­ous with the endoplasmic reticulum and ribo­somes are often attached to the external surface.

Pneumocystosis, Lung, Rat 219

Intracystic bodies develop into extracystic troph­oblasts. Empty cysts are irregular in shape and often ap­pear crescentic because of collapse of the wall.

Differential Diagnosis

Since the intraalveolar accumulations of Pneumo­cystis appear honeycombed and contain tropho­zoite nuclei, this lesion can be easily differentiated from pulmonary edema, which often accompa­nies it and may be the primary finding after inha­lation of toxic substances. Depending on the im­mune defect present, a small number of inflam­matory cells, usually macrophages, may be found. Alveolar wall infiltration is usually slight; how­ever, in the presence of associated bacterial or fungal infection it may be severe. Pneumocystis must be differentiated from other organisms. Their nuclei stain with hematoxylin, but are less densely stained than most gram-posi­tive bacteria, which also stain with hematoxylin. Cyst walls stained with silver, methyl violet, or to­luidine blue must be differentiated from fungi, es­pecially Candida (Tornlopsis) glabrata (Fig. 270). The finding of crescentic or cup-shaped cysts is characteristic of Pneumocystis, as is the irregular wrinkling of the intact cyst. Budding would be characteristic for yeast; Candida glabrata, which is the most common source of confusion, can be cultured. The eight-nucleated cyst should always be searched for in Giemsa-stained imprints for confirmation of the diagnosis (Fig. 271).

Biologic Features

Natural History. Most colonies of rats are natural­ly infected, as are some colonies of mice and rab­bits. Normally infections are subclinical and ia­trogenic immunosuppression, usually with corti­costeroids or cyclophosphamide, or profound malnutrition are necessary to give rise to clinically significant pulmonary involvement (Frenkel et al. 1966; Frenkel 1976; Robbins et al. 1976). Athymic nude mice are also susceptible (Walzer and Rut­ledge 1980). Spontaneous instances of pneumo­cystosis have been described in cats, horses, goats, pigs, dogs, hares, owl monkeys, chimpanzees, and others (Chandler et al. 1976). The presence of significant pulmonary involve­ment with Pneumocystis is an indication of im­munosuppression. Hughes (1982) showed that

Page 230: Respiratory System

220 1. K. Frenkel

Page 231: Respiratory System

naturally infected lung tissue is not infectious and that rats become infected from contaminated air. The infectious stage has not yet been determined.

Pathogenesis. In normal rats pneumocystis organ­isms are quite scarce and the Delanoes, who described and named Pneumocystis, reported searching three or four impression smears before finding a single cyst in normal rats (Delanoe and Delanoe 1912). In athymic rats, small focal infil­trates may be found. Only after cortisone became available was massive pulmonary involvement produced in rats (Frenkel et al. 1966; Chandler et al. 1979). Earlier workers encountered Pneumo­cystis accompanied by pneumonia and abscesses from Corynebacterium kutscheri, staphylococci, Pseudomonas, Proteus, Streptobacillus monilifor­mis, Pasteurella pneumotropica, coliforms, and fungi. The use of chlortetracycline in drinking wa­ter (50 mg/dl) and of amphotericin B, 1 mg three times weekly subcutaneously, prevented most bacterial and fungal infections and permitted the study of relatively pure Pneumocystis infection. The following other regimes have led to clinical pneumocystosis in 200 g, 6-8 week old rats: 25 mg cortisone acetate given subcutaneously twice weekly alternating with cortisol; dexamethasone sodium phosphate, 1 mg/l in drinking water; placing 250 g rats on 8% protein diet; cyclophos-

<I Fig.265 (Upper left). Rat lung. Most alveoli filled with masses of Pneumocystis carinii (P); trapped air in some alveoli (A). The bronchi (B) contain a small amount of fluid, devoid of Pneumocystis. PAS and H, x 80

Fig.266 (Upper right). Alveolar filling with Pneumocystis. Rat was treated with 25 mg cortisone acetate twice weekly, subcutaneously, for 64 days. With hematoxylin and eosin, the organisms appear as faintly hematoxylin-staining nu­clei in a foamy eosinophilic material. A few inflammatory macrophages are present. Hand E, x 630

Fig.267 (Lower left). Same lung as in Fig. 266. With the PAS-hematoxylin technique, the foamy matrix is empha­sized, but the hematoxylin-staining nuclei can also be seen. PAS and H, x 630

Fig.268 (Lower right). Same lung as in Fig.266.With the Grocott methenamine silver technique, the cysts resemble spherical or collapsed yeasts, such as Histoplasma or Toru­lopsis;this technique is useful mainly to identify candidate forms which must be confirmed by Giemsa staining in smears; some collagen and reticulum fibers are also pre­sent. Grocott methenamine silver, x 630

Pneumocystosis, Lung, Rat 221

Fig.269. Pneumocystis carinii cysts in section of lung im­pregnated with silver appear as flattened, wrinkled, or folded yeast-like bodies. Grocott methenamine silver, x 1500

phamide, 240 mg/kg body weight orally three times weeky; aminopterin and chlorambucil together with subeffective doses of cortisone acetate; and cyclosporin A 10 mg/kg per day orally. Masses of Pneumocystis tachyzoites fill the alveo­li, leading to anoxia in immunosuppressed hosts (Fig. 266). Ordinarily, little inflammatory reaction is present, in part because the organism is not highly chemotactic and, in part, because the im­munosuppressive state is accompanied by an im­paired inflammatory response. Inflammation ap­pears when immunocompetence returns, for ex­ample when corticosteroid administration is stopped. Inflammation also appears when the or­ganisms are killed by the administration of pent­amidine, or by sulfonamide and a dihydrofolate reductase inhibitor (Frenkel et al. 1966; Hughes et al. 1974), even though corticosteroids are still ad­ministered. All of these causes of resurgent in­flammatory reaction may initially result in further

Page 232: Respiratory System

222 1. K. Frenkel

<1 Fig. 270 (Above). Fungal yeasts. This strain (of Candida (Toru/opsis) glabrata) has ovoid singular nuclei and would not easily be confused with Pneumocystis, but round yeasts could be easily confused. Smear from culture. Giemsa, x 1500

Fig.271 (Below). A Pneumocystis cyst in impression smear with eight-nucleated interior bodies (center). Numerous Pneumocystis trophozoites in the periphery. A distinct nu­cleus and cytoplasm can be seen. Giemsa, x 1500

impaired alveolocapillary diffusion and increased anoxia, until some of the Pneumocystis are elimi­nated. The organisms are essentially confined to the alveoli, where they are closely attached to type I pneumocytes, which later degenerate (Walzer et al. 1980; Yoneda and Walzer 1981). They are sometimes phagocytized and may be carried to hilar lymph nodes, but have not been reported to multiply there in laboratory animals. Because of their tight attachment to type I pneumocytes, the organisms are not expectorated and are not found in the bronchi, except after the adminis­tration of chemotherapy with sulfadiazine-pyri­methamine, sulfamethoxazole-trimethoprim, or pentamidine.

Etiology. Carlos Chagas first recognized the eight­nucleated cysts in lungs of guinea pigs infected with Trypanosoma entzi and, believing it to indi­cate schizogony and to be part of the trypanosom­al cycle, created the genus Schizotrypanum (Cha­gas 1909). Pneumocystis carinii was described as a separate organism by Delanoe and Delanoe (1912) in the lungs of rats and later in guinea pigs from Paris. The organism had generally been re­garded as a protozoan, although in the absence of characteristic organelles, its affinity to protozoan orders remains obscure. On the basis of the study of its ultrastructure, Vavra and Kucera (1970) stressed the similarity of this organism to fungi. Although the cyst resembles a fungus, the ame­boid trophozoites do not appear to, being further­more devoid of chitin, although other polysaccha­rides are present. The susceptibility of Pneumocys­tis to pentamidine is shared by two protozoa, Leishmania and Trypanosoma, and by the fungus Histoplasma; however, the resistance of Pneumo­cystis to amphotericin B makes a fungal classifica­tion less likely, although some fungi are resistant. It is likely that the taxonomic affinities of the agent will become clearer once the infectious stage has been identified.

Page 233: Respiratory System

Frequency. All conventional rats examined in 1964 and 1965 and even five or six lines advertised as "specific-pathogen-free" (SPF) developed pneu­mocystosis when treated with cortisone acetate, 25 mg twice weekly: one SPF line and one ex­germ-free line, recently contaminated with bacte­ria, remained free of Pneumocystis at the same time.

Comparison with Other Species

Pneumocystosis in man was recognized in institu­tionalized premature babies and in infants at the end of World War II in Europe. The histologic picture was of an interstitial plasma cell pneumo­nia with masses of Pneumocystis filling the alveoli. Plasma cell pneumonia has not been observed in experimental animals. In an immunosuppressed baby with congenital rubella, both plasma cells and large mononuclear cells are present. How­ever, no significant interstitial inflammation, as in the experimental models, is found in children with agammaglobulinemia due to deficiency of B cells (Bruton's disease), in patients with leukemia or lymphoma treated with appropriate chemo­therapy, or in organ transplant recipients immu­nosuppressed with corticosteroids and azathio­prine. Because there are serological differences between P. carinii of rats, the human organism was designated Pneumocystis jiroveci (Frenkel 1976). Athymic nude mice are sometimes observed with focal Pneumocystis accumulations; cortisone­treated rabbits and mice are also found to be in­fected (Sheldon 1959; Walzer et al. 1980). Recently, pneumocystosis has been described in humans with the acquired immunodeficiency syn­drome and in nonhuman primates which were im­munosuppressed for unknown reasons (Chandler et al. 1976).

Pneumocystosis, Lung, Rat 223

References

Campbell WG JR (1972) Ultrastructure of Pneumocystis in human lung. Arch Pathol93: 312-324

Chagas C (1909) Nova tripanozomiaza humana. Estudos sobre a morfolojia e 0 cicio evolutivo de Schizotrypanum crnzi n.gen., n. sp. ajenta etiologio de nova entidade morbida de homan. Mem Inst Oswaldo Cruz 1 : 159-164

Chandler FW, McClure HM, Campbell WG Jr, Watts JC (1976) Pulmonary pneumocystosis in nonhuman pri­mates. Arch Pathol Lab Med 100: 163-167

Chandler FW Jr, FrenkeUK, Campbell WG Jr (1979) Pneumocystis pneumonia. Animal model: Pneumocystis carinii pneumonia in the immunosuppressed rat. Am J PathoI95:571-574

De1anoe P, Delanoe Mme (1912) Sur les rapports des kystes de Carini du poumon des rats avec Ie Trypanoso­ma lewisii. C. R. Sequces Acad Sci 155: 658-664

FrenkeUK (1976) Pneumocystis jiroveci n. sp. from man: morphology, physiology, and immunology in relation to pathology. Natl Cancer Inst Monogr 43: 13-30

Frenkel JK, Good JT, Shultz JA (1966) Latent Pneumocys­tis infection of rats, relapse and chemotherapy. Lab In­vest 15: 1559-1577

Hughes WT (1982) Natural mode of acquisition for de no­vo infection with Pneumocystis carinii. J Infect Dis 145: 842-848

Hughes WT, McNabb OC, Makres TD, Feldman S (1974) Efficacy of trimethoprim and sulfamethoxazole in the prevention and treatment of Pneumocystis carinii pneumonitis. Antimicrob Agents Chern other 5: 289-293

Macher A, Shelhamer J, Parker M, Parrillo J, Gill V, Ma­surH (1983) Emergency open lung biopsy at NIH: use of a new modified methylene blue stain for the rapid tis­sue demonstration of Legionella, Pneumocystis, cytome­galovirus, Nocardia and fungi causing opportunistic pneumonias. Crit Care Med 11: 221 (abstract)

RobbinsJB, DeVitaVT Jr, DutzW (eds) (1976) Sympo­sium on Pneumocystis cariniiinfection. Natl Cancer Inst Monogr43

Sheldon WH (1959) Experimental pulmonary Pneumo­cystis carinii infection in rabbits. J Exp Med 110: 147-160

VavraJ, Kucera K (1970) Pneumocystis carinii delanoe, its ultrastructure and ultrastructural affinities. J Protozool 17: 463-483

Walzer PD, Rutledge ME (1980) Comparison of rat, mouse, and human Pneumocystis carinii by immunoflu­orescence. J Infect Dis 142: 449

Walzer PD, Powell RD Jr, Yoneda K, Rutledge ME, Mil­der JE (1980) Growth characteristics and pathogenesis of experimental Pneumocystis carinii pneumonia. Infect Immun 27: 928-937

Yoneda K, Walzer PD (1981) Mechanism of pulmonary alveolar injury in experimental Pneumocystis carinii pneumonia in the rat. Br J Exp Pathol62: 339-346

Page 234: Respiratory System

224 1. K. Frenkel

Aspergillosis and Mucormycosis, Lung, Rat

J. K. Frenkel

Synonyms. None (aspergillosis); phycomycosis, zygomycosis, hyphomycosis (mucormycosis).

Gross Appearance

Members of fungal species give rise to focal and diffuse pneumonia, which often progresses to ne­crosis, vascular invasion and infarction in immu­nosuppressed rats, and occasionally in mice and hamsters. The affected areas are raised with white, necrotic centers which are surrounded by gray, consolidated tissue and hyperemic borders (Fig. 272). Fibrinous pleuritis usually accompa­nies the lesions.

Microscopic Features

Diffuse pneumonia is accompanied by fungal hy­phae, generally with tissue necrosis and infarc-

Fig. 272. Rat lung with two white plaques (arrows) covering the pleural surface. The rat was treated with 25 mg corti­sone acetate subcutaneously, twice weekly for 64 days

tion. The fungi may exhibit nonseptate hyphae, characteristic of Mucor, or septate hyphae, com­patible with Aspergillus. Fungal bronchitis is com­mon with Candida, a fungal yeast, but Aspergillus and Mucor also give rise to bronchitis. Aspergillus infection next to a bronchus may erode into it, forming an ulcer, a cavity, and sometimes a fungal ball. Growth of the fungus may occur through the pleura into the pleural sac (Figs. 273 and 274). The cellular exudate contains mononuclear cells and granulocytes, which occasionally leads to granuloma formation. Aspergillus hyphae are of approximately constant width, are septate, and often undergo dichotomous branching. In the presence of a bronchus or a cavity, conidia (fruit­ing bodies) may be formed. The agent of Mucor has hyphae of greatly varied width which are sparsely septate with haphazard branching; the hyphae sometimes appear twisted or folded, mis­takenly suggesting septation (Fig. 275). Vascular invasion is common and blood vessels often serve as conduits for the spread of the fungi.

Ultrastructure

The ultrastructure is rarely examined because the organisms are readily visible by light microscopy and their identification depends on culture. The fungal hyphae appear as tubules with prominent cell walls.

Differential Diagnosis

Fungal granulomas, abscesses, and pneumonias must be differentiated from bacterial abscesses and pneumonias using impression smears, histo­logic sections, and cultures. Fungal and bacterial infections may both be present, sometimes even with viral infections and Pneumocystis, a protozo­an, in immunosuppressed animals. Because fun­gal contaminants are so common, one cannot rely on culture alone, and the isolated organisms must be shown by evidence in histologic sections to be related to the lesion.

Page 235: Respiratory System

Biologic Features

Natural History. The fungi are widely distributed in the environment, especially in moist feed and bedding (Emmons 1962), and the spores are readily inhaled by animals. Immunocompetent animals control the infection, which remains sub­clinical; occasionally, a single aspergilloma may be found, sometimes with cavitation. Not so in the immunosuppressed animal, where focal fungal bronchitis leads to necrotizing bronchopneumo­nia, vascular invasion, dissemination in the lung and occasionally to the other organs, and exten­sion to the pleura. Thrombi form in the infected vessels and may give rise to infarcts without fungi, which are mainly aerobic.

Pathogenesis. In both aspergillosis and mucormy­cosis, tissue invasion from the bronchi and vascu­lar invasion are predominant. It is likely that pro­teolytic enzymes favor the fungi's penetration through the connective tissue; collagenases and elastases have been isolated from some of the Mu­corales. Acidosis markedly lowers natural resis­tance of hosts to mucormycosis, as has been ob­served clinically in humans, usually those with diabetes. This was studied in rabbits that had re­ceived injections of alloxan (Sheldon and Bauer 1962). Acidosis temporarily decreased the levels of an inhibitory serum factor. Athymic mice ap­pear to be resistant (Corbel and Eades 1977), but corticosteroids markedly depress the capacity to react to these fungi (Frenkel 1962). Endotoxin, hemolysins, and other factors from these fungi have been described but do not appear to play an obvious role in the pathogenesis of clinical illness. Aflatoxin, however, causes acute liver necrosis and scarring in many animals, the effective dose varying according to species; it is also a potent carcinogen, again with wide variations in sensitiv­ity according to species and according to hormo­nal states (Goodall and Butler 1969).

Etiology. Several species of Aspergillus (A.jlavus, A. niger, A.fumigatus) and species of Mucor, Absi­dia, Rhizopus, and Mortierella may be found in laboratory animals. The diagnosis is often diffi­cult because some of the Mucorales do not grow easily in culture. Experimental infections with A.fumigatus, A.jlavus, A. niger, and Rhizopus ory­zae are commonly fatal.

Frequency. Infections are rare in normal animals unless they are exposed to very large doses of fun­gi, as from moist and moldy feed. The frequency

Aspergillosis and Mucormycosis, Lung, Rat 225

Fig.273 (Above). Lung with fungal hyphae growing from parenchyma into pleura (F). Some of the nearby blood ves­sels contain similar fungi, but the bronchi are free. PAS and H, x 25

Fig.274 (Below). Aspergillus terreus (identified by culture) in pleura of rat. Septate fungal hyphae of constant bore and with occasional spores in pleura (P) and subpleural area. PAS and H, x 400

Page 236: Respiratory System

226 J. K. Frenkel

Fig.275. Mucormycosis, lung, rat. Nonseptate fungal hy­phae of irregular diameter. Blood vessels were also in­volved, turning this area of lung into a hemorrhagic infarct. This fungus was not cultured. PAS and H, x 400

of illness is directly related to the degree of im­munosuppression. The infection can be sup­pressed by the administration of amphotericin B, 6 mg/kg body weight (rat) administered intramus­cularly three times weekly (Frenkel et al. 1966).

Comparison with Other Species

The tissue lesions described are quite similar in rats and humans. Mice and hamsters appear more resistant than rats. Aspergillus bronchitis is com­mon in certain birds, leading often to disseminat­ed infection. Allergic bronchitis due to fungi is rare in experimental animals but does occur in man. Mucorales have been reported to give rise to illness in animals fed moist and moldy food, with generalized infection common at the time of death (Jones and Hunt 1983). The rhino facial mu­cormycosis of humans has also been seen in a rhe­sus monkey (Martin et al. 1969), but appears to be rare in small laboratory animals.

References

Corbel MJ, Eades SM (1977) Experimental mucormycosis in congenitally athymic (nude) mice. Mycopathologia 62:117-120

Emmons CW (1962) Natural occurrence of opportunistic fungi. Lab Invest 11: 1026-1032

Frenkel JK (1962) Role of corticosteroids as predisposing factors in fungal diseases. Lab Invest 11: 1192-1208

Frenke1JK, Good JT, Shultz J A (1966) Latent Pneumocys­tis infection of rats, relapse and chemotherapy. Lab In­vest 15: 1559-1577

Goodall CM, Butler WH (1969) Aflatoxin carcinogenesis: inhibition of liver cancer induction in hypophysecto­mized rats. Int J Cancer 4: 422-429

Jones TC, Hunt RD (1983) Veterinary pathology, 5th edn. Lea and Febiger, Philadelphia

Martin JE, Kroe DJ, Bostrom RE, Johnson DJ, Whit­ney RA Jr (1969) Rhino-orbital phycomycosis in a rhe­sus monkey (Macaca mulatta). J Am Vet Med Assoc 155:1253-1257

Sheldon WH, Bauer H (1962) The role of predisposing fac­tors in experimental fungus infections. Lab Invest 11: 1184-1191

Page 237: Respiratory System

Toxoplasmosis, Lung, Mouse, and Hamster 227

Toxoplasmosis, Lung, Mouse and Hamster

J. K. Frenkel

Synonyms. None.

Gross Appearance

Focal to confluent pneumonias, often with serous pleural exudate, are seen at necropsy of animals which succumb to acute infection. In immuno­suppressed animals in which a chronic infection relapses, white to yellowish necrotic foci sur­rounded by hemorrhage are seen in the lungs in addition to diffuse pneumonia.

Microscopic Features

Toxoplasma tachyzoites, the rapidly multiplying forms, are found in the alveolar walls, in cells of alveolar exudate, and in many other cells of the body (Fig. 27 6). Diffuse interstitial pneumonia is the usual picture. More rarely, focal necrosis in­volves segments of the alveolar wall. Intracellular and free tachyzoites, a mixed mononuclear cell infiltration, and serofibrinous exudate accom­pany both (Fig.277). In immunosuppressed ani­mals, larger focal lesions involving several alveoli many be found, with diffuse cellular infiltration and necrosis or early organization by fibroblasts (Fig. 278). Focal fibrinous pleuritis is present with parenchymal lesions reaching the pleura. Rarely, Toxoplasma cysts containing bradyzoites or slow­ly multiplying forms are found in the lungs with­out inflammatory reaction.

Ultrastructure

The ultrastructure of Toxoplasma is useful to dis­tinguish it from some other small organisms that may be encountered in the lungs. Toxoplasma tachyzoites are banana-shaped to ovoid and carry an apical complex at the anterior end composed of a conoid, a polar ring, and rhoptries, which contain enzymes that apparently aid their active penetration of host cells. Also, a vesicular nucle­us, mitochondria, endoplasmic reticulum, micro­tubules, and micronemes are enclosed in a pellicle (Chobotar and Scholtyseck 1982).

Differential Diagnosis

The apical complex is present in other Sporozoa, such as intestinal and cyst-forming coccidia, which has led to a redefinition of the phylum as the Apicomplexa. Members of this group need to be differentiated by their cycle and cyst structure. However, because of their small size they must al­so be distinguished from Leishmania spp. and Trypanosoma cruzi, which have a kinetoplast; from Encephalitozoon, which contains a spiral po­lar filament; from Pneumocystis, which lacks of a special organelle; and from Histoplasma and oth­er fungal yeasts with their PAS-positive cell wall.

Biologic Features

Natural History. Toxoplasma gondiiis an intestinal coccidian of cats with an enteroepithelial cycle that leads to the development of oocysts that are shed in the feces. Oocysts sporulate outside the host. Ground-feeding animals and herbivores be­come infected by ingesting vegetation mixed with soil contaminated with cat feces. These animals constitute the intermediate host; all mammals and birds investigated have been found to be suscepti­ble. During acute infection tachyzoites multiply actively, giving rise to necrosis of infected cells. Coincident with the development of immunity, bradyzoites slowly develop within tissue cysts, with cells of the brain, muscle and other organs supporting large numbers of organisms. The cysts appear in the chronic, persisting stage of infec­tion, and when eaten by a carnivore, produce ta­chyzoites. Only in felines will they initiate an in­traepithelial and a sexual cycle in the gut. Laboratory animals are likely to become infected by the ingestion of oocysts from either contami­nated food or bedding. Cats are known to cover their feces and will defecate sometimes in open sacks or bins containing feed and cage bedding (Fig. 279).

Pathogenesis. The tachyzoites of acute infection multiply by successive division (endodiogeny), reaching eight to 32 organisms per cell, at which time the cell usually disintegrates and the tachyzo­ites infect other cells. Bradyzoites develop in mice 4-5 days after infection with tachyzoites, 7-9 days

Page 238: Respiratory System

228 J. K. Frenkel

Fig.276 (Upper left). Focal toxoplasmic pneumonia, ham­ster (Fig.278) with tachyzoites in parenchyma, bronchial epithelium, and exudate. Groups of tachyzoites (arrows). Hand E, x 450

Fig.277 (Upper right). Toxoplasmic pneumonia. Nude mouse with partially functioning thymic transplant, died 22 days after subcutaneous inoculation with Toxoplasma tachyzoites. Group oftachyzoites (arrow). Hand E, x 400

Fig.278 (Below). Focal toxoplasmic pneumonia. Syrian golden hamster latently infected with Toxoplasma for several months. Immunosuppression with subcutaneously administered cortisol, 2.5 mg twice weekly, was then be­gun. Forty-five days later this animal died with focal pneu­monia and focal encephalitis. Hand E, x 25

Page 239: Respiratory System

Toxoplasmosis, Lung, Mouse, and Hamster 229

HAY GRAIN

FECAL CONTAM.

MEAT ••••••••

'-"

Fig. 279. Transmission of toxoplasmosis in zoos and animal colonies.

after infection with bradyzoites, and 9-10 days af­ter infection with sporozoites from occysts. The slow multiplication of bradyzoites leads to accumulations of tens to hundreds of organisms within intracellular cysts. These cysts contain or­ganisms in resting stages, awaiting ingestion by a carnivore, and have little pathogenicity to the host cell, which maintains the cyst for months to years. However, disintegration of a cyst may lead to tis­sue necrosis in the presence of delayed hypersen­sitivity (type IV); this is of clinical importance when cyst rupture occurs in the retina, because function is concentrated in a few cells which do not regenerate. The liberated organisms are usual­ly destroyed by immune mechanisms. Minimal pathogenicity is also associated with the prolifera­tion of stages A, B, C, D, and E and the develop­ment of gametocytes in the intestinal epithelial cells of cats, because it coincides more or less with the natural turnover of epithelial cells which ma­ture while migrating from the crypts to the tips of the villi where they are shed.

Etiology. Toxoplasma gondii was originally de­scribed as a Leishmania or related organism in 1908, both in North Africa and South America. Nicolle and Manceaux found it in gondis, wild rodents used in studies of Leishmania and typhus

at the Pasteur Institute in Tunis. These animals presumably became infected in the laboratory. Splendore found Toxoplasma in laboratory rab­bits in San Paulo, Brazil. The absence of a kineto­plast separated it from Leishmania. In 1909 it was named Toxoplasma gondii by Nicolle and Man­ceaux (reviewed by Frenkel 1973). The organism was suspected to give rise to human disease spo­radically, but it was not definitely linked with hu­man disease until Wolf, Cowan, and Paige, start­ing in 1937, identified Toxoplasma in newborn babies in New York, isolated it by 1940, and linked it to intrauterine infection from an asymp­tomatic mother. At first the organism was called Encephalitozoon, as in a case described by lanku in 1923 (Frenkel 1973). But by 1940, when the or­ganism had been isolated from an infected baby, it was recognized by Sabin as identical with Toxo­plasma isolated from animals (Frenkel 1973). This was followed by characterization of the nednatal infection by Sabin, the development of serologic tests, especially the dye test by Sabin and Feld­man, and the recognition of ocular, encephalitic, and lymph node syndromes, apart from general­ized infection in children and adults. Toxoplasma was found to give rise to disease in many domes­tic, wild, zoo, and laboratory animals, without, be­cause of its size and number, causing serious er-

Page 240: Respiratory System

230 J. K. Frenkel

rors in the interpretation of laboratory experi­ments (Frenkel 1973). Although it was realized that carnivorism could b~ responsible for transmitting toxoplasmosis, it did not become clear how herbivores became in­fected till 1965, when W. Hutchison discovered the infectivity of the feces of a cat that had eaten Toxoplasma-infected mice. In 1967 he described the supposed transmission in nematode eggs. Be­tween 1969 and 1970 transmission without the nematode was established and a small Toxoplas­ma oocyst was recognized in cat feces (reviewed by Frenkel 1973). Toxoplasma is now classified in the protozoan phylum Apicomplexa, as a cyst­forming, heteroxenous coccidian in the family Sarcocystidae, subfamily Toxoplasmatina (Fren­kel1977). Toxoplasma differs from the genera Be­snoitia, Hammondia, Cystoisospora, Sarcocystis, and Frenkelia by the characteristics of the cysts and the transmission cycle (Frenkel 1973, 1977; Frenkel et al. 1979).

Frequency. Naturally occurring toxoplasmosis with illness and sometimes death has been ob­served in gondis, rabbits, guinea pigs, neotropical primates, and pigeons. Asymptomatic infection has been observed in guinea pigs, rats, chickens, and goats. Cats used to be given a free run in some animal colonies to catch wild and acciden­tally escaped mice. Disposition of the feces of these cats was not considered important, and it is likely that some of the feces were deposited in grain bins and bedding. With improved practices in handling animal feed and bedding and exclu­sion of pet and stray cats, the opportunity for in­fection can be diminished. Experimental toxo­plasI?osis has been widely studied in mice, rabbits, and hamsters: these animals are more sus­ceptible than rats and guinea pigs, which develop an age-dependent resistance. In these laboratory studies, direct transmission does not occur from animal to animal except by occasional cannibal­ism and congenital infection. However, experi­mental infection with oocysts from cat feces must be conducted with care, because a portion of the fed oocysts may be passed intact in the feces, where they will withstand drying and are thus ca­pable of infecting other animals. Bedding should be changed and autoclaved 1 day and 2 days after the administration of oocysts. Food and bedding are the fomites, there is no evidence of airborne infection.

Comparison with Other Species

Generalized toxoplasmosis is similar in animals and man, keeping in mind their different degrees of resistance and strains of Toxoplasma which dif­fer in pathogenicity. Well-adapted laboratory strains may convey a distorted impression, killing mice 3-4 days after intraperitoneal inoculation of a large number of organisms. However, of 31 re­cent natural isolates, 90% gave rise to inapparent infections in mice (Ruiz and Frenkel 1980) and fa­tal infections could be transformed to chronic in­fections when mice were treated prophylactically with 15-60 mg% of sulfadiazine or sulfamerazine in the drinking water. Adult rats are resistant even to strains pathogenic to mice, as are adult guinea pigs, adult humans, and most children. Ocular toxoplasmosis in man has been reproduced in hamsters; after subcutaneous inoculation, fol­lowed by sulfadiazine treatment, the retinas of many of these animals became infected (Frenkel 1961). Relapsing chronic toxoplasmosis seen in humans has also been reproduced in hamsters treated with pharmacologic doses of cortico­steroids or cyclophosphamide (Frenkel et al. 1978). Most experimental infections are produced by injection, the intraperitoneal route being much more effective than the subcutaneous. Oral infec­tion is the usual route in animals and man, either with oocysts from cat feces or with tissue cysts from infected meat (Dubey and Frenkel 1973). The course of the disease and the development of immunity may differ according to the route of in­fection.

References

Chobotar B, Scholtyseck E (1982) Ultrastructure. In: Long PL (ed) The biology of the coccidia. University Park Press, Baltimore, chap 4

Dubey JP, Frenkel JK (1973) Experimental toxoplasma in­fection in mice with strains producing oocysts. J Parasi­to159: 505-512

FrenkeIJK (1961) Pathogenesis of toxoplasmosis with a consideration of cyst rupture in Besnoitia infection. Surv Ophthalmol6: 799-825

Frenkel JK (1973) Toxoplasma in and around us. Bio­science 23: 343-352

FrenkeIJK (1977) Bensoitia wallacei of cats and rodents: with a reclassification of other cyst-forming isosporoid coccidia. J Parasitol 63: 611-628

Frenkel JK, Amare M, Larsen W (1978) Immune compe­tence in a patient with Hodgkin's disease and relapsing toxoplasmosis. Infection 6 (2): 84-91

FrenkelJK, HeydornAO, Mehlhorn H, Rommel M (1979) Sarcocystinae: Nomina dubia and available names. Z Parasitenkd 58: 115-139

Ruiz A, FrenkeIJK (1980) Intermediate and transport hosts of Toxoplasma gondii in Costa Rica. Am J Trop Med Hyg 29: 1161-1166

Page 241: Respiratory System

Subject Index*

A, mice, 106t mouse strain, 148t

Acatalasemic, mouse, 150t Acetate, cortisone, 218, 220f Acetoxypropylnitrosamine, APPN, 28t ACI N, rat, 115t Acrylate, ethyl, 46t Adenocarcinoma, 47, 50f, 112

anterior nasal epithelium rat, 67 ethmoid region, 49f, 50f ethomoturbinal, 48f lung hamster, 132t

mouse metastasis renal, 145f maxilloturbinal, 67f nasoturbinal, 68f type B, lung mouse metastatic

mammary, 143f Adenoma, 41

alveologenic, 99,102 bronchiolo-alveolar, 102, 108 cell, type II, l04f Clara cell, 108f, 108, 110f lung hamster, 132t

mouse, alveolar type II cell, 102 bronchiolar, 108 type II, 102f, 103f, 104f

cell, 105f rat, bronchiolar alveolar, 99, 100f

maxilloturbinate rat, polypoid, 43f nasal cavity rat, 46t

mucosa rat, polypoid, 41 nonciliated light cell, 45f nose rat, polypoid, 42f papillary, 41, 108, 110f, Illf polypoid,41f pulmonary, 99,102 rat, polypoid nasal, 44f type II, 103f

cell,102 Adenomatosis, bronchioalveolar, 177

pulmonary, 177 Adenomatous polyp, 33, 41 Adenosquamos carcinoma lung

hamster, 132t Aeruginosa, Pseudomonas, 215 AlBrA mouse strain, 148t Alkylnitrosamines, 122 Alveolar cell carcinoma, 112

rat, type II, 96f epithelialization, 177 exudate bleomycin, lung mouse,

161f histiocytosis rat, 169

hyperplasia, 177 lipoproteinosis, lung rat, 173f, 174f

rat, 171 lung, 171f, 172f

macrophage, asbestos body, 184f proteinosis, 171 squamous metaplasia, 199f type II cell adenoma lung mouse,

102 Alveoli hamster lung, 96f Alveologenic adenoma, 99,102

carcinoma lung mouse, 146f Alveolus bleomycin, fibrosis lung

mouse, 163f giant cells hair particles, 191f hair particles pulmonary, 191f

Amelanotic melanoma mouse, pulmonary metastasis, 139f

Anaplastic carcinoma lung hamster, 120f

Anatomy nasal cavity rat, 3 Angiectasis,74t Anterior nasal cavity, squamous cell

carcinoma, 56f epithelium rat, adenocarcinoma,

67 Anthracosis, 180 APPN acetoxypropylnitrosamine, 28t APUD-type cells, neuroendocrine,

121f Arm, dynein, 92f Aromatic hydrocarbons, halogenated,

201 PAHs Polycyclic, 28t

Artery embolic hair, pulmonary, 189f lung mouse mammary tumor emboli

pulmonary, 141f thrombotic pulmonary, 187f

Asbestos bodies, 185f body, 186f

alveolar macrophage, 184f cholesterol clefts, 185f inhalation, 184f pneumoconiosis, 183

Asbestosis hamster, 183 Ash hamster, fly, 182t

pneumoconiosis hamster, fly, 180 Aspergillosis mucormycosis lung rat,

224 rat lung, 224f

Aspergillus terreus pleura rat, 225f Auratus, Mesocricetus, 33 Axon, intraepithelial, 8f

B6C3 Fl mice, 74t endogenous lipid pneumonia, 166

mouse, 148t, 149t BaA Benz(a)anthracene, 28t Baboon, pulmonary toxicity bleomycin, 165t

BALB/c C, mice, 106t C3H/He mouse strain, 148t cfC3H/Cbl Se mouse strain, 148t cfC3H mouse strain, 148t CfRIII mouse strain, 148t cNIV mouse strain, 148t cStCrl, mouse, 149t, 150t

BaP Benzo(a)pyrene, 28t N-Nitrosobis acetoxypropyl amine, 28t

Basal lamina, 8f Benz(a)anthracene, BaA, 28t Benzo(a)pyrene, 122

BaP,28t Benzo(e)pyrene, BeP, 28t BeP Benzo e pyrene, 28t Besnoitia, 230 BHP, 63f, 65f, 122

N-Nitrosobis hydroxypropyl amine, 28t

Biochemistry hypophysectomized rat, serum,169t

Bladder, metastasis lung urinary, 150t Bleeding, tail veins, 188t, 193t Bleomycin baboon, pulmonary toxicity, 165t

dog, pulmonary toxicity, 165t fibrosis lung mouse, 162f

alveolus, 163f hamster, pulmonary toxicity, 165t injury mouse pulmonary fibrosis, 160

lung mouse alveolar exudate, 161f pulmonary blood vessels, 162f vascular lesion, 161f

mouse, pulmonary toxicity, 165t pheasant, pulmonary toxicity, 165t

Blood vessels bleomycin, lung mouse pulmonary, 162f

Bodies, asbestos, 185f Body alveolar macrophage, asbeStos,

184f . asbestos, 186f giant cells, foreign, 189f granuloma foreign, 189f hamster lung, neuroepithelial, 93f

Bordetella bronchiseptica, 215

* Note: Page numbers in boldface indicate the principal discussion; figures are designated by the letter "P' following the page number; tables are found on page numbers followed by the letter "t"

Page 242: Respiratory System

232 Subject Index

Bronchial epithelium, serous cell rat, 90f

lymphoid tissue rat, normal, 215f Bronchiectasis, enzootic, 213 Bronchioalveolar adenomatosis, 177 Bronchiolar adenoma lung mouse, 108

alveolar adenoma lung rat, 99, 100f carcinoma lung rat, 112, 114f

rat,l13f hyperplasia F344 rat, 179t

lung rat, 177f, 177, 178f tumors rats, naturally occuring,

115t carcinoma, 112 epithelium, proliferation, 185f metaplasia, 177

Bronchiole multiple epithelial syncytia, 196f

murine respiratory mycoplasmosis, 216f

rat lung brush cell, 94f Clara cell, 94f

Bronchiolization lung hamster, 181f Bronchiolo-alveolar adenoma, 102, 108

Bronchiseptica, Bordetella, 215 Bronchus hamster, 121f

murine respiratory mycoplasmosis, 216f

Brush cell, 7f, 9f bronchiole rat lung, 94f

Bulla purulent exudate, tympanic, 80f

C3H/Cb/Se mouse strain, 148t He mouse strain, BALB/C, 148t mice,106t mouse strain, 148t

C3HeB/FEJ hybrid, mouse, 149t C57BLl6J mouse strain, 148t C57 black, mice, 106t

leaden L or M, mice, 106t C57BL/6J mouse, squamous cell carcinoma pulmonary vein, 130f

mouse strain, 148t C57BLl6Jx, mouse, 149t Candida turolopsis glabrata, 219, 222f Carcinogens respiratory epithelium

syrian golden hamster, 33 Carcinoma, 112

alveolar cell, 112 anterior nasal cavity, squamous cell,

56f bronchiolar, 112, epidermoid, 54, 62, 117 in situ, dysplasia, 34 larynx, clear cell, 75f, 76f

syrian hamster, clear cell, 75 lung F344 rat, squamous cell, 129f

hamster, adenosquamous, 132t anaplastic, 120f squamous cell, 119f, 132t

mouse, alveologenic, 146f hepatocellular, 148t metastasis harderian gland,

145f

renal transitional cell, 145f metastatic hepatocellular, 143f

rat, bronchiolar alveolar, 112, 114f epidermoid, 124, 127 naturally occurring squamous

cell,126t radiation-induced squamous

cell, 127 squamous cell, 124p, 125f, 128f

syrian hamster, squamous cell, 117 mouse mammary gland, 148t nasal,60f

cavity rat, squamous cell, 58f mucosa rat, squamous cell, 54, 59t

squamous cell, 55f nasoturbinate rat, squamous cell, 57f pulmonary vein C57Bl J mouse, squamous cell, 130f

rat, bronchiolar alveolar, 113f lung, squamous cell, 124 mouse hamster man, comparison

squamous cell, 132t squamous cell, 47, 63f, 64f, 65f upper respiratory tract syrian

hamster, squamous ce, 62 Carinii, Pneumocystis, 221f, 223

rat lung Pneumocystis, 220f Catarrh, infections, 78 Caudal vein male rat, thrombus, 190f CBA mouse strain, 148t Cell adenoma, Clara, 108f, 108, 110f

lung mouse, alveolar type II, 102 type II, 105f

nonciliated light, 45f type II, 102

bronchiole rat lung brush, 94f Clara,94f

brush, 7f, 9f carcinoma, alveolar, 112

anterior nasal cavity, squamous, 56f

larynx, clear, 75f, 76f syrian hamster, clear, 75

lung F344 rat, squamous, 129f hamster squamous, 119f, 132t mouse metastasis renal transitional, 145f

rat, naturally occurring squamous, 126t

radiation-induced squamous, 127

squamous, 124, 125f, 128f syrian hamster, squamous, 117

nasal cavity rat, squamous, 58f mucosa rat, squamous, 54p, 59t

squamous, 55f nasoturbinate rat, squamous, 57f pulmonary vein C57Bl J mouse,

squamous, 130f rat lung, squamous, 124

mouse hamster man, comparison squamous, 132t

squamous, 47, 63f, 64f, 65f upper respiratory tract syrian

hamster, squamo, 62

ciliated, 9f, 20f Clara,111f granuloma hair particle, lung giant,

193t lung giant, 188t, 193t skin particle, lung giant, 193t

hyperplasia, goblet, 30 mucous,30f

lung mouse type II epithelial, 163f Mycoplasma pulmonis, tracheal epithelial, 82f

nonciliated columnar, 7f, 9f dark,44f

papilloma, squamous, 33 pneumonia, foamy, 166

interstitial plasma, 218 rat bronchial epithelium, serous, 90f

type II alveolar, 96f secretory, 26f tumor, lung mouse metastasis

ovarian granulosa, 144f squamous,62,117 type I, 14f, 24f, 25f

II, 14f, 19f adenoma, 104f ciliated, 16f, 18f

Cells, cuboidal, 7f differentiation ciliated, 17

mucous, 26 mucous, 20

epithelial, 12f foreign body giant, 189f hair particles alveolus, giant, 191f hair-phagocytosing giant, 192t malignant squamous, 64f, 65f, 118f neuroendocrine APUD-type, 121f type I mucous, 22f

CF-l, mouse, 149t, 150t strain, 148t

Chemically induced lung tumors F344 rats,101t

Chloride, dimethy1carbamoyl, 59t hydrogen, 59t

Chlormethyl ether, 59t Cholesterol clefts asbestos, 185f

granulomas, lung rat, 174f pneumonia, 166

CHR chrysene, 28t Chronic murine pneumonia, 213

respiratory mycoplasmosis, 81f respiratory disease, murine, 78, 213

Crysene, CHR, 28t Ciliated cell, 9f, 20f

type II, 16f, 18f cells, differentiation, 17 mucous cells, differentiation, 26

Cilium, human respiratory, 92f Clara cell, ll1f

adenoma, 108, 108f, 110f bronchiole rat lung, 94f

Clear cell carcinoma larynx, 75f, 76f syrian hamster, 75

Clefts asbestos, cholesterol, 185f Coliforms,221 Columnar cell, nonciliated, 7f, 9f

Page 243: Respiratory System

Comparison squamous cell carcinoma rat mouse hamster man, 132t

Coronaviridae, 86, 212 Coronavirus infection, 86

lung, rat, 204f rat, 203

Parker's rat, 203 interstitial pneumonia rat, 204f

Coronaviruses, mouse, 201 Corticosteroids, 219, 230 Cortisone acetate, 218, 220f Corynebacterium kutscheri, 215, 221

mouse, 201 rat, 201

Cruzi, Trypanosoma, 227 Cs, mouse, 150t Cuboidal cells, 7f Cyclophosphamide, 219, 230 Cyst, pneumocystis, 222f Cystoisopora, 230

Dark cell, nonciliated, 44f Dba, mice, 106t

mouse strain, 148t xC57BL Fl, mouse, 149t

DBF1, mouse, 149t DBN,122

N-Nitrosodibuthylamine, 28t N-nitrosodiethylamine diethyl­nitrosamine, 28t

Desquamative interstitial pneumonia, 171

pneumonia, 171 DHPN Dihydroxy-di-n-propylnitrosamine, 28t

Diagnosis papillary tumors, differential, 39

Dibromo-3-chloropropane,51f Dibromochloropropane, 67f Dibromoethane,101t Diethylnitrosamine, DEN

N-nitrosodiethylamine,28t Differential diagnosis papillary tumors, 39

Differentiation, 13 ciliated cells, 17

mucous cells, 26 mucous cells, 20

Dihydroxy-di-n-propylnitrosamine, DHPN,28t

Dimethyl sulfoxide, DMSO, 28t Dimethyl-l benzanthracene, 122

DMBA,28t Dimethylcarbamoyl chloride, 59t Dinitrosopiperazine, 46t Dioxane, 59t Disease, murine chronic respiratory, 78,213

DMBA 9,1 O-Dimethyl-l ,2-benzanthracene, 28t

DMDPN N-Nitrosobis (2-methylpropyl) amine, 28t

DMSO Dimethyl sulfoxide, 28t Dog, pulmonary toxicity bleomycin, 165t

DPN,64f N-Nitrosodi-n-propylamine,28t

Duct, nasolacrimal, 4f Dynein arm, 92f Dysplasia carcinoma in situ, 34

trachea, 35f Dysplastic epithelium, 29f

squamous epithelium, 63f

Emboli lung, skin, 186 pulmonary artery, lung mouse

mammary tumor, 141f vascular system, hair fragment,

186 Embolic hair, 187f

fragment lung, 193f pulmonary artery, 189f

Embolism hairs, pulmonary, 186 pulmonary hair, 186

Encephalitozoon, 227 Endogenous lipid pneomonia, 171

B6C3Fl mice, 166 mouse, 166f, 167f

Enzootic bronchiectasis, 213 Epichlorohydrin, 59t Epidermoid carcinoma, 54, 62, 117

lung rat, 124, 127 metaplasia, 30 papillary tumor, 33

Epithelia, olfactory, 6f respiratory, 6f

Epithelial cell, lung mouse type II, 163f Mycoplasma pulmonis, tracheal, 82f

cells,12f papilloma, 33 syncytia, bronchiole multiple, 196f

Epithelialization, alveolar, 177 Epithelioid mesothelioma hamster, 134f, 135f, 136f

Epithelium, dysplastic, 29f squamous, 63f

hamster, tracheal, l1f olfactory, 9f proliferation bronchiolar, 185f rat, adenocarcinoma anterior nasal, 67

trachea, 89f respiratory, 6f, 7f, 8f, 9f, 36 serous cell rat bronchial, 90f syrian golden hamster, carcinogens

respiratory, 33 tracheal, 11

tracheal, 13f, 15f, 21f, 24f Esthesioneuroblastoma, 47 Ether, chlormethyl, 59t

phenylglycidyl, 59t Ethmoid region, adenocarcinoma, 49f,

50f turbinate, 4f turbinates rat, neoplasms mucosa,

47 Ethomoturbinal, adenocarcinoma, 48f Ethyl acrylate, 46t Explant culture fetal trachea, 28

Subject Index 233

cultures fetal trachea syrian golden hamsters, 27

fetal trachea, 30f tracheal, 29f

Exudate bleomycin, lung mouse alveolar,161f tympanic bulla purulent, 80f

F344, rat, 115t, 126t bronchiolar alveolar hyperplasia,

179t olfactory region, 52f, 53f squamous cell carcinoma lung,

129f rats, chemically induced lung tumors,10H

FC3H/Nctr, mouse, 149t, 150t Fetal trachea, 30f

explant, 30f culture, 28

syrian golden hamsters, explant cultures, 27

tracheal explant, 29f Fibrosarcoma lung mouse, 13H

rat, 131t, 131t Fibrosis, bleomycin injury mouse

pulmonary, 160 lung mouse alveolus bleomycin,

163f bleomycin, 162f

Fly ash hamster, 182t pneumoconiosis hamster, 180

Foamy cell pneumonia, 166 Foreign body giant cells, 189f

granuloma, 189f Formaldehyde, 46t, 57f, 59t, 59t

nasoturbinate rat, 43f Frenkelia, 230 Frequency pulmonary tumors mice, 106t

Function lung, structure, 89 Fungal hyphae, lung, 225f

yeasts, 222f Fungi,221 Fusocellular mesothelioma hamster, 135f

Gases, oxidant, 201 Giant cell granuloma hair particle, lung, 193t

lung, 188t, 193t skin particle, lung, 193t

cells, foreign body, 189f hair particles alveolus, 191f hair-phagocytosing, 192t

Giardia, mouse, 201 Glabrata, Candida torulopsis, 219,

222f Gland carcinoma, lung mouse

metastasis harderian, 145f mouse mammary, 148t

metastasis lung harderian, 150t mammary,15Ot

virus infection, rat submaxillary, 84, 210

Page 244: Respiratory System

234 Subject Index

Glands rat trachea, seromucous, 91f Goblet cell hyperplasia, 30 Golden hamster, carcinogens respira­

tory epithelium syrian, 33 tracheal epithelium, syrian, 11

hamsters, explant cultures fetal trachea syrian, 27

Gondii, Toxoplasma, 227, 229 GR mouse strain, 148t Granuloma foreign body, 189f

hair particle, lung giant cell, 193t lung giant cell, 188t, 193t skin particle, lung giant cell, 193t

Granulomas, lung rat cholesterol, 174f Granulosa cell tumor, lung mouse

metastasis ovarian, 144f

Hair, embolic, 187f embolism, pulmonary, 186 fragment emboli pulmonary

vascular system, 186 lung, embolic, 193f

intravenous injection, 191f particle, lung giant cell granuloma,

193t thromboarteritis, 193t

particles alveolus, giant cells, 191 f lung, 188t pulmonary alveolus, 191f

pulmonary artery embolic, 189f segment, pigmented, 187f tail veins thrombophlebitis, 193t thromboarteritis, tail veins, 188t vessel, tail veins, 193t

Hair-phagocytosing giant cells, 192t Hairs, pulmonary embolism, 186 Halogenated aromatic hydrocarbons,

201 Hammondia, 230 Hamster, adenocarcinoma lung, 132t

adenoma lung, 132t adenosquamous carcinoma lung,

132t Anaplastic carcinoma lung, 120f asbestosis, 183 bronchiolization lung, 181f bronchus, 121f carcinogens respiratory epithelium

syrian golden, 33 clear cell carcinoma larynx syrian, 75

epithelioid mesothelioma, 134f, 135f,136f

fly ash, 182t pneumoconiosis, 180

fusocellular mesothelioma, 135f hyperplasia lung, 181f lung, alveoli, 96f

interalveolar septum, 96f neuroepithelial body, 93f

man, comparison squamous cell carcinoma rat mouse, 132t

papillary mesothelioma, 134f mucoepidermoid tumor trachea, 33f

peripheral lung, 95f pleural mesothelioma syrian, 133 pneumoconiosis lung, 180f pulmonary toxicity bleomycin, 165t radioactive materials, lung tumor

syrian, 132t squamous cell carcinoma, lung,

119f,132t syrian, 117

upper respiratory tract syri, 62

metaplasia, lung, l18f toxoplasmic pneumonia, 228f toxoplasmosis lung mouse, 227 trachea, 22f tracheal epithelium, 11 f

syrian golden, 11 Hamsters, explant cultures fetal trachea syrian golden, 27

Harderian gland carcinoma, lung mouse metastasis, 145f

metastasis lung, 150t Hemagglutinating virus infection JHV, Japanese, 195

mouse HVM, 195 Japan HVJ, 195

Hemangioma,74t Hemangiosarcoma,74t

lung mouse, 131t rat,131t

nasal cavity, 73f, 74f mouse, 72

Hepatoblastoma, lung mouse, 148t Hepatocellular carcinoma, lung mouse, 148t

metastatic, 143f Hepatoma, lung mouse, 148t HEPES 4 (2-Hydroxyethyl)-1-piperazineethane sulfonic acid, 28t

Hexamethylphosphoramide, 46t, 59t Histiocytosis, multifocal, 171

rat, alveolar, 169 Histoplasma, 227 Holtzman-SD, rat, 115t HPPN N-Nitroso-2-hydroxypropyl-n­

propylamine, 28t Human respiratory cilium, 92f HVJ, hemagglutinating virus Japan,

195 HVM, hemagglutinating virus infection mouse, 195

Hybrid, mouse C3HeB FEJ, 149t Hydrocarbons, halogenated aromatic, 201

PAHs Polycyclic aromatic, 28t Hydrogen chloride, 59t Hydroxyethyl piperazineethane sulfonic acid, HEPES, 28t

Hyperplasia, 34 alveolar, 177 F344 rat, bronchiolar alveolar, 179t goblet cell, 30 lung hamster, 181f

rat, bronchiolar alveolar, 177f, 177, 178f

mucous cell, 30f trachea,35f

Hyphae, lung fungal, 225f Hyphomycosis, 224 Hypophysectomized rat, serum biochemistry, 169t

I, mice, 106t Induced tumors mice, pulmonary

metastasis, 149t Infection, coronavirus, 86

JHV, Japanese hemagglutinating virus, 195

lesions due to, 195 lung mouse rat, pneumonia virus

mice, 206 sendai virus, 195

sialodacryoadenitis virus, 210 rat coronavirus, 204f

rat coronavirus, 203 mouse HVM, hemagglutinating virus, 195

parainfluenza virus, 195 Parkers rat coronavirus, 203 rat submaxillary gland virus, 84, 210 reparative phase, sendai virus, 197f resolution phase, sendai virus, 200f SDAV, 84, 210 sendai virus, 196f, 197f upper respiratory tract rat, sialo­

dacryoadenitis vi, 84 Infections, 78

catarrh, 78 Inhalation asbestos, 184f Injection hair, intravenous, 191f Injury mouse pulmonary fibrosis, bleomycin, 160

Interalveolar septum hamster lung, 96f Interstitial plasma cell pneumonia, 218

pneumonia, desquamative, 171 PVM rat, 208f rat coronavirus, 204f

Intraepithelial axon, 8f Intravenous injection hair, 1911'

Japan HVJ, hemagglutinating virus, 195 Japanese hemagglutinating virus

infection JHV, 195 JHV, Japanese hemagglutinating virus

infection, 195 Jiroveci, Pneumocystis, 223

K virus, mouse, 201 Klebsielle pneumoniae, 215 Kutscheri, Corynebacterium,/215, 221

mouse Corynebacterium, 201 rat Corynebacterium, 201

Lamina, basal, 8f Larynx, clear cell carcinoma, 75f, 76f

syrian hamster, clear cell carcinoma, 75

Leishmania spp, 227 Lesion bleomycin, lung mouse vascular, 161f

Page 245: Respiratory System

Lesions due to infection, 195 lung, nonneoplastic, 160 microembolic pulmonary, 186 tail veins lung, 188t

Sprague-Dawley rat, 193t Light cell adenoma, nonciliated, 45f Lipid pneumonia B6C3Fl mice, endogenous, 166

endogenous, 171 mouse, endogenous, 166f, 167f

Lipidosis rat, pulmonary, 169t, 169, 170f

Lipoproteinosis, lung rat alveolar, 173f, 174f

rat, alveolar, 171 lung alveolar, 171f, 172f

Liver, metastasis lung, 149t Lung, 87

alveolar lipoproteinosis rat, 171f, 172f

alveoli hamster, 96f aspergillosis, rat, 224f brush cell, bronchiole rat, 94f Clara cell, bronchiole rat, 94f embolic hair fragment, 193f F344 rat, squamous cell carcinoma, 129f

fungal hyphae, 225f giant cell granuloma, 188t, 193t

hair particle, 193t skin particle, 193t

hair particles, 188t hamster, adenocarcinoma, 132t

adenoma, 132t adenosquamos carcinoma, 132t anaplastic carcinoma, 120f bronchiolization,181f hyperplasia, 181f peripheral,95f pneumoconiosis, 180f squamous cell carcinoma, 119f,

132t metaplasia, 118f

harderian gland, metastasis, 150t interalveolar septum hamster, 96f lesions tail veins, 188t liver, metastasis, 149t mammary gland, metastasis, 150t metastasis, 138 mouse alveolar exudate bleomycin,

161f type II cell adenoma, 102

alveologenic carcinoma, 146f alveolus bleomycin, fibrosis, 163f bleomycin, fibrosis, 162f bronchiolar adenoma, 108 fibrosarcoma, 131 t hamster, toxoplasmosis, 227 hemangiosarcoma, 1311 hepatoblastoma, 148t hepatocellular carcinoma, 148t hepatoma, 148t lymphosarcoma,131t mammary tumor emboli pulmonary artery, 141f

metastasis harderian gland carcinoma, 145f

malignant schwannoma, 146f ovarian granulosa cell tumor,

144f renal adenocarcinoma, 145f

transitional cell carcinoma, 145f

subcutaneous sarcoma, 141f metastatic hepatocellular

carcinoma, 143f mammary adenocarcinoma type B, 143f

tumors, 138 pleural metastasis osteosarcoma,

146f pulmonary blood vessels bleomycin, 162f

rat, pneumonia virus mice infection, 206

sendai virus infection, 195 sialodacryoadenitis virus

infection, 210 thrombi malignant schwannoma,

146f type II adenoma, 102f, 103f, 104f

cell adenoma, 105f epithelial cell, 163f

vascular lesion bleomycin, 161f neuroepithelial body hamster, 93f nonneoplastic lesions, 160 ovary, metastasis, 150t Pneumocystis carinii, rat, 220f pneumonia virus mice rat, 207f rat alveolar lipoproteinosis, 173f,

174f aspergillosis mucormycosis, 224 bronchiolar alveolar adenoma, 99,

100f carcinoma, 112, 114f hyperplasia, 177f, 177, 178f

cholesterol granulomas, 174f coronavirus infection, 204f epidermoid carcinoma, 124, 127 fibrosarcoma, 131t, 131t hemangiosarcoma,131t lymphosarcoma,131t mesothelioma, 131 t mucormycosis, 226f murine respiratory

mycoplasmosis, 213 mycoplasmosis,215f naturally occurring squamous cell

carcinoma, 126t normal, 214f pneumocystosis, 218f, 218 radiation-induced squamous cell

carcinoma, 127 rat coronavirus infection, 203 reticulosarcoma,131t squamous cell carcinoma, 124,

125f,128f RIll mouse metastatic mammary tumor, 140f

secondary tumors, 138

Subject Index 235

skin emboli, 186 metastasis, 150t particles, 188t

spleen, metastasis, 150t Sprague-Dawley rat, lesions tail veins, 193t

squamous cell carcinoma rat, 124 structure function, 89 syrian hamster, squamous cell

carcinoma, 117 thromboarteritis, 188t, 193t

hair particle, 193t skin particle, 193t

tumor mice radioactive materials, 131t

rat, 1311 syrian hamster radioactive

materials, 132t tumors F344 rats, chemically

induced, 1011 urinary bladder, metastasis, 150t

Lymphoid tissue rat, normal bronchial, 215f

Lymphosarcoma lung mouse, 131t rat,131t

M-2-0B N-Nitrosomethyl (2-oxobu­tyl) amine, 28t

Macrophage, asbestos body alveolar, 184f

Male rat, thrombus caudal vein, 190f Malignant mesothelioma, 133

schwannoma, lung mouse metastasis, 146f

thrombi,146f squamous cells, 64f, 65f, 118f

Mammary adenocarcinoma type B, lung mouse metastatic, 143f

gland carcinoma, mouse, 148t metastasis lung, 150t

tumor,138f emboli pulmonary artery, lung

mouse,141f lung, RIll mouse metastatic, 140f

Man, comparison squamous cell carcinoma rat mouse hamster, 132t

Mass, rat subcutaneous, 55f Materials, lung tumor mice radioactive, 1311

syrian hamster radioactive, 132t radioactive,131t

Maxilloturbinal, adenocarcinoma, 67f Maxilloturbinate, 4f

rat, polypoid adenoma, 43f Melanoma mouse, pulmonary metastasis amelanotic, 139f

pulmonary metastasis melanotic, 139f

Melanttic melanoma mouse, pulmonary metastasis, 139f

Mesocricetus auratus, 33 Mesothelial neoplasia, 133 Mesothelioma hamster, epithelioid,

134f, 135f, 136f fusocellular, 135f

Page 246: Respiratory System

236 Subject Index

Mesothelioma hamster papillary, 134f

lung rat, 131t malignant, 133 syrian hamster, pleural, 133

Metaplasia, alveolar squamous, 199f bronchiolar, 177 epidermoid, 30 lung hamster squamous, 118f simple, 35 squamous, 29f, 30f, 34, 36

Metastasis amelanotic melanoma mouse, pulmonary, 139f harderian gland carcinoma, lung

mouse, 145f induced tumors mice, pulmonary,

149t lung, 138

harderian gland, 150t liver, 149t mammary gland, 150t ovary,150t skin, 150t spleen, 150t urinary bladder, 150t

malignant schwannoma, lung mouse, 146f

melanotic melanoma mouse, pulmonary, 139f

osteosarcoma, lung mouse pleural, 146f

ovarian granulosa cell tumor, lung mouse,l44f

renal adenocarcinoma, lung mouse, 145f transitional cell carcinoma, lung

mouse, 145f subcutaneous sarcoma, lung mouse,

141f untreated mice, pulmonary, 148t

Metastatic hepatocellular carcinoma, lung mouse, 143f mammary adenocarcinoma type B, lung mouse, 143f tumor lung, RIll mouse, 140f

tumors lung mouse, 138 Methylnitrosourea, MNU, 28t Mice A, 106t

B6C3Fl,74t BALBI c C, 106t C3H,106t C57 black, 106t

leaden, Lor M, 106t dba,106t endogenous lipid pneumonia

B6C3F1,166 frequency pulmonary tumors, 106t 1,106t infection lung mouse rat,

pneumonia virus, 206 pneumonia virus, 209 pulmonary metastasis induced tumors, 149t

untreated 148t PVM, pneumonia virus, 206

radioactive materials, lung tumor, 131t

rat lung, pneumonia virus, 207f Swiss,106t

Microembolic pulmonary lesions, 186 Mixed polyp, 33 MNU Methylnitrosourea, 28t Moniliformis, Streptobacillus, 215, 221 MOP N-Nitrosomethyl oxopropyl amine,28t

Mouse acatalasemic, 150t alveolar exudate bleomycin, lung,

161f type II cell adenoma lung, 102

alveologenic carcinoma lung, 146f alveolus bleomycin, fibrosis lung,

163f B6C3F1,149t BALB/cStCr1, 149t, 150t bleomycin, fibrosis lung, 162f bronchiolar adenoma lung, 108 C3HeB/FEJ hybrid, 149t C57BLl6Jx,149t CF-l, 149t, 150t coronariruses, 201 Corynebacterium kutscheri, 201 Csb, 150t DBAxC57BLlF1,149t DBF1,149t endogenous lipid pneumonia, 166f,

167f fC3H Nctr, 149t, 150t fibrosarcoma lung, 131t giardia, 201 hamster man, comparison squa­

mous cell carcinoma rat, 132t toxoplasmosis lung, 227

hemangiosarcoma lung, 131t nasal cavity, 72

hepatoblastoma, lung, 148t hepatocellular carcinoma, lung, 148t hepatoma, lung, 148t HVM, hemagglutinating virus infection, 195

K virus, 201 lymphosarcoma lung, 131t mammary gland carcinoma, 148t

tumor emboli pulmonary artery, lung, 141f

metastasis harderian gland carcinoma, lung, 145f

malignant schwannoma, lung, 146f

ovarian granulosa cell tumor, lung,l44f

renal adenocarcinoma, lung, 145f transitional cell carcinoma,

lung, 145f subcutaneous sarcoma, lung, 141f

metastatic hepatocellular carcinoma, lung, 143f mammary adenocarcinoma type

B, lung, 143f tumor lung, RIll, 140f

tumors lung, 138

Mycoplasma pulmonis, 201 pleural metastasis osteosarcoma,

lung, 146f pneumocystis, 201 pneumonia virus, 201, 206 pulmonary metastasis amelanotic melanoma, 139f

pulmonary blood vessels bleomycin, lung, 162f fibrosis, bleomycin injury, 160 metastasis melanotic melanoma,

139f toxicity bleomycin, 165t

rat, pneumonia virus mice infection lung, 206

sendai virus infection lung, 195 sialodacryoadenitis virus infection

lung, 210 spironucleus, 201 squamous cell carcinoma pulmo­

nary vein C57Bl J, 130f strain, A, 148t, 148t

AlBrA,148t B6C3HF1, 148t BALB/C/C3H/He,148t

cfC3H/Cb/Se,148t cfC3H,148t cfRIII,148t cNIV,148t

C3H/HE,148t Cb/Se,148t

C57BLl6J,148t CBA,148t Cf-l,148t DBA, 148t OR, 148t NIH white, 148t RIll, 148t

Dm/Se,148t thrombi malignant schwannoma,

lung, 146f toxoplasma, 201 toxoplasmic pneumonia nude, 228f type II adenoma lung, 102f, 103f,

104f cell adenoma lung, 105f epithelial cell, lung, 163f

vascular lesion bleomycin, lung, 161f

MPN N- Nitrosomethyl-n-propyl­amine,28t

Muco-epidermoid papillary tumor, 36f Mucoepidermoid papillary tumor, 33

respiratory tumor, 39f tumor trachea hamster, pal?illary, 33f

Mucormycosis lung rat, 226£ aspergillosis, 224

Mucosa ethmoid turbinates rat, neoplasms, 47

normal nasal septal, 79f rat, polypoid adenoma nasal, 41

squamous cell carcinoma nasal, 54,59t

tracheal, 81f Mucous cell hyperplasia, 30f

Page 247: Respiratory System

cells, differentiation, 20 ciliated, 26

type J, 22f Multifocal histiocytosis, 171 Multiple epithelial syncytia, bronchiole,196f

Murine chronic respiratory, disease, 78,213

pneumonia, chronic, 213 respiratory mycoplasmosis, 79f

bronchiole, 216f bronchus,216f chronic,81f lung rat, 213 rat, 213f, 214f

mycoplasmosis upper respiratory tract rat, 78

Mycoplasma pulmonalis, 215 pulmonis, 78, 82, 217

mouse, 201 rat, 201 tracheal epithelial cell, 82f

Mycoplasmosis, bronchiole murine respiratory,216f bronchus murine respiratory, 216f chronic murine respiratory, 81f lung rat, 215f

murine respiratory, 213 murine respiratory, 79f rat, murine respiratory, 213f, 214f

Mycoplasmosis upper respiratory tract rat, murine respiratory, 78

N-6-MI N-Nitrosohexamethyle­imine,28t

N-nitroso-2-hydroxypropyl-n-propyla­mine, 122

HPPN,28t N -Nitrose-2-oxopropyl-n-propyl­amine, OPPN, 28t

N-nitrosobis (2-acetoxypropyl) amine, 122

N-Nitrosobis acetoxypropyl amine, BAP,28t (2-hydroxypropyl) amine, 122

BHP,28t (2-methylpropyl) amine, DMDPN,

28t N-nitrosodi-n-propylamine, 122

DPN,28t N-Nitrosodibuthylamine, DBN, 28t N-nitrosodiethylamine, 121f

diethylnitrosamine, DEN, 28t N-nitrosohexamethyleneimine,122

N-6-MI,28t N-Nitrosomethyl (2-oxobutyl) amine,

M-2-0B,28t (2-oxopropyl)amine,122

MOP,28t N-nitrosomethyl-n-propylamine, 122

MPN,28t N-nitrosomethylpiperazine, 49f, 52f N-Nitrosomorpholine, NM, 28t N-nitrosovinylethylamine, 122

YEN,28t

Nasal adenoma rat, polypoid, 44f carcinoma, 60f cavity, hemangiosarcoma, 73f, 74f

mouse, hemangiosarcoma, 72 rat,3f

adenoma, 46t anatomy, 3 squamous cell carcimona, 58f

squamous cell carcinoma anterior, 56f

epithelium rat, adenocarcinoma anterior, 67

mucosa rat, polypoid adenoma, 41 squamous cell carcinoma, 54,

59t squamous cell carcinoma, 55f

septal mucosa, normal, 79f Nasolacrimal duct, 4f Nasopharynx,4f Nasoturbinal, adenocarcinoma, 68f Nasoturbinate,4f

rat formaldehyde, 43f squamous cell carcinoma, 57f

Naturally occurring bronchiolar alveolar tumors rats, 115t

squamous cell carcinoma lung rat, 126t

Neoplasia, mesothelial, 133 Neoplasms, 33

mucosa ethmoid turbinates rat, 47 Neuroblastoma, olfactory, 47 Neuroendocrine APUD-type cells, 121f

Neuroepithelial body hamster lung, 93f

NIH white mouse strain, 148t Nitroacenaphthene, 101 t Nitrosamines, 63 Nitrosaminobutanone, 46t Nitrosomethylurea, NMU, 28t NM N-Nitrosomorpholine, 28t NMU Nitrosomethylurea, 28t Nonciliated columnar cell, 7f, 9f

dark cell, 44f light cell adenoma, 45f

Nonneoplastic lesions lung, 160 Normal bronchial lymphoid tissue rat, 215f

lung rat, 214f nasal septal mucosa, 79f rat trachea, 81f

Nose rat, polypoid adenoma, 42f Nude mouse, toxoplasmic pneumonia, 228f

Olfactory epithelia, 6f epithelium, 9f neuroblastoma, 47 organ, septal, 4f region F344 rat, 52f, 53f

tumor,51f OPPN,65f

N -Nitroso-2-oxopropyl-n-propy­lam,28t

Oregon, rat, 115t

Subject Index 237

Organ, septal olfactory, 4f vomeronasal, 4f

Osborne-Mendel, rat, 115t, 126t Osteosarcoma, lung mouse pleural

metastasis, 146f Ovarian granulosa cell tumor, lung

mouse metastasis, 144f Ovary, metastasis lung, 150t Oxidant gases, 201 Oxide, propylene, 74t

P-cresidine, 46t, 50f, 59t PAHs Polycyclic aromatic hydrocarbons, 28t

Papillary adenoma, 41, 108, 11 Of, ll1f mesothelioma hamster, 134f mucoepidermoid tumor trachea

hamster, 33f polyp, 33 tumor, epidermoid, 33

muco-epidermoid, 36f mucoepidermoid, 33 trachea,37f

tumors, 34 differential diagnosis, 39 ultrastructure, 37

Papilloma, 33, 41 epithelial, 33 squamous cell, 33

Parainfluenza virus, 202 infection, 195

Paramyxovirus, 202 Parker's rat coronavirus infection, 203 Particles alveolus, giant cells hair, 191f

lung hair, 188t skin, 188t

pulmonary alveolus, hair, 191f Pasteurella pneumotropica, 215, 221 Peripheral lung hamster, 95f Periphlebitis, tail veins, 188t, 193t Pheasant, pulmonary toxicity bleomycin, 165t

Phenacetin, 46t, 59t Phenylglycidyl ether, 59t Phlebitis, tail veins, 188t Phycomycosis, 224 Pigmented hair segment, 187f Plasma cell pneumonia, interstitial, 218 Pleura rat, Aspergillus terreus, 225f Pleural mesothelioma syrian hamster, 133

metastasis osteosarcoma, lung mouse, 146f

Pneumoconiosis, 180 asbestos, 183 hamster, fly ash, 180 lung hamster, 180f

Pneumocystis, 219, 227 carinii, 221f, 223

rat lung, 220f cyst, 222f jiroveci, 223 mouse, 201 trophozoites, 222f

Pneumocystosis lung rat, 218f, 218

Page 248: Respiratory System

238 Subject Index

Pneumonia B6C3Fl mice, endogenous lipid, 166

cholesterol, 166 chronic murine, 213 desquamative, 171

interstitial, 171 endogenous lipid, 171 foamy cell, 166 hamster, toxoplasmic, 228f interstitial plasma cell, 218 nouse, endogenous lipid, 166f, 167f nude mouse, toxoplasmic, 228f PVM rat, interstitial, 208f rat coronavirus, interstitial, 204f sialodacryoadenitis viral, 211 f virus, 206

mice, 209 infection lung mouse rat, 206 PVM,206 rat lung, 207f

mouse, 201, 206 Pneumoniae, Klebsielle, 215

rat Streptococcus, 201 Streptococcus, 215

Pneumotropica, Pasteurella, 215, 221

Pneumovirus, 209 Polycyclic aromatic hydrocarbons,

PAHs,28t Polyp, 33

adenomatous, 33, 41 mixed, 33 papillary, 33

Polypoid adenoma, 41f maxilloturbinate rat, 43f nasal mucosa rat, 41 nose rat, 42f

nasal adenoma rat, 44f tumor, 33

Predifferentiation stage, 11 Proliferation bronchiolar epithelium,

185f Propylene oxide, 74t Proteinosis, alveolar, 171 Proteus, 221 Pseudomonas, 221

aeruginosa,215 Pulmonary metastasis amelanotic melanoma mouse, 139f

Pulmonalis, Mycoplasma, 215 Pulmonary adenoma, 99,102

adenomatosis, 177 alveolus, hair particles, 191f artery embolic hair, 189f

lung mouse mammary tumor emboli, 141f

thrombotic, 187f blood vessels bleomycin, lung

mouse, 162f embolism hairs, 186 fibrosis, bleomycin injury mouse, 160

hair embolism, 186 lesions, microembolic, 186 lipidosis rat, 196t, 169, 170f

metastasis induced tumors mice, 149t

melanotic melanoma mouse, 139f untreated mice, 148t

toxicity bleomycin baboon, 165t dog, 165t hamster, 165t mouse, 165t pheasant, 165t

tumors mice, frequency, 106t vascular system, hair fragment

emboli, 186 vein C57BL/6J mouse, squamous

cell carcinoma, 130f Pulmonis, mouse Mycoplasma, 201

Mycoplasma, 78, 82, 217 rat Mycoplasma, 201 tracheal epithelial cell Mycoplas­

ma,82f Purulent exudate, tympanic bulla, 80f PVM, pneumonia virus mice, 206

rat, interstitial pneumonia, 208f

Radiation-induced squamous cell carcinoma lung rat, 127

Radioactive materials, 131t lung tumor mice, 131t

syrian hamster, 132t Rat ACIIN, 115t

adenocarcinoma anterior nasal epithelium, 67

adenoma nasal cavity, 46t alveolar histiocytosis, 169

lipoproteinosis,171 lung, 173f, 174f

anatomy nasal cavity, 3 aspergillosis mucormycosis lung,

224 Aspergillus terreus pleura, 225f bronchial epithelium, serous cell, 90f bronchiolar alveolar adenoma lung, 99,100f

carcinoma,113f lung, 112, 114f

hyperplasia F344, 179t lung, 177, 177~ 178f

cholesterol granulomas, lung, 174f coronavirus infection lung, 204f

rat, 203 Parker's, 203

interstitial pneumonia, 204f Corynebacterium kutscheri, 201 epidermoid carcinoma lung, 124,

127 F344, 115t, 126t fibrosarcoma lung, 131t, 131t formaldehyde, nasoturbinate, 43f hemangiosarcoma lung, 131t Holtzman-SO, 115 interstitial pneumonia PVM, 208f lesions tail veins lung

Sprague-Dawley, 193t lung alveolar lipoproteinosis, 171f,

172f aspergillosis, 224f

brush cell, bronchiole, 94f Clara cell, bronchiole, 94f Pneumocystis carinii, 220f pneumonia virus mice, 207f squamous cell carcinoma, 124 tumor,131t

lymphosarcoma lung, 131t mesothelioma lung, 131t mouse hamster man, comparison squamous cell carcinoma, 132t

mucormycosis lung, 226f murine respiratory mycoplasmosis, 213f,214f

lung, 213 mycoplasmosis upper respira-tory tract, 78

Mycoplasma pUlmonis, 201 mycoplasmosis lung, 215f nasal cavity, 3f naturally occurring squamous cell

carcinoma lung, 126t neoplasms mucosa ethmoid turbinates, 47

normal bronchial lymphoid tissue, 215f

lung, 214f olfactory region F344, 53f Oregon, 115t Osborne-Mendel, 115t, 126t pneumocystosis lung, 218, 218f pneumonia virus mice infection lung

mouse, 206 polypoid adenoma maxilloturbinate,43f

nasal mucosa, 41 nose,42f

nasaladenoma,44f pulmonary lipidosis, 169t, 169, 170f radiation-induced squamous cell

carcinoma lung, 127 rat coronavirus infection lung, 203 reticulosarcoma lung, 131t sendai virus infection lung mouse,

195 serum biochemistry hypophysec­

tomized, 169t Sherman, 115t sialodacryoadenitis virus infection

upper respiratory tr, 84 Sprague-Dawley, 115t

Crl COPS (SO), 115t HAP (SO), 115t

squamous cell carcinoma lung, 124, 125f,128f

F344, 129f nasal cavity, 58f

mucosa, 54, 59t nasoturbinate, 57f

Streptococcus pneumoniae, 201 subcutaneous mass, 55f submaxillary gland virus infection,

84,210 thrombus caudal vein male, 190f trachea, epithelium, 89f

normal,81f

Page 249: Respiratory System

seromucous glands, 91f type II alveolar cell, 96f ventral turbinate, 85f Wistar, 115t, 126t

Rats, chemically induced lung tumors F344,101t naturally occurring bronchiolar alveolar tumors, 115t

Renal adenocarcinoma, lung mouse metastasis, 145f transitional cell carcinoma, lung

mouse metastasis, 145f Reparative phase, sendai virus infection, 197f

Resolution phase, sendai virus infection, 200f

Respiratory cilium, human, 92f disease, murine chronic, 78, 213 epithelia, 6f epithelium, 6f, 7f, 8f, 9f, 36

syrian golden hamster, carcinogens, 33

mycoplasmosis, bronchiole murine, 216f bronchus murine, 216f chronic murine, 81 f lung rat, murine, 213 murine,79f rat, murine, 213f, 214f

mycoplasmosis upper respiratory tract rat, murine, 78

syncytial virus, RSV, 209 system, upper, 1 tract rat, murine respiratory mycoplasmosis upper, 78

sialodacryoadenitis virus infection up, 84

syrian hamster, squamous cell carcinoma upp, 62

tumor, mucoepidermoid, 39f Reticulosarcoma lung rat, 131t RIIl/Dm/Se mouse strain, 148t

mouse metastatic mammary tumor lung, 140f

strain, 148t RSV, respiratory syncytial virus, 209

Sarcocystis, 230 Sarcoma, lung mouse metastasis sub­

cutaneous, 141f Schwannoma, lung mouse metastasis

malignant, 146f thrombi malignant, 146f

SDAV infection, 84, 210 sialodacryoadenitis virus, 210

Secondary tumors lung, 138 Secretory cell, 26f Segment, pigmented hair, 187f Sendai virus infection, 196f, 197f

lung mouse rat, 195 reparative phase, 197f resolution phase, 200f

Septal mucosa, normal nasal, 79f olfactory organ, 4f

Septum hamster lung, interalveolar, 96f

Seromucous glands rat trachea, 91f Serous cell rat bronchial epithelium,

90f Serum biochemistry

hypophysectomized rat, 169t Sherman, rat, 115t Sialodacryoadenitis, 84f, 85f

viral pneumonia, 211f virus, 86

infection lung mouse, 210 upper respiratory tract, 84

SDAV,210 Simple metaplasia, 35 Skin emboli lung, 186

metastasis lung, 150t particle, lung giant cell granuloma,

193t thromboarteritis, 193t

particles, lung, 188t Spironucleus, mouse, 201 Spleen, metastasis lung, 150t Spp, Leishmania, 227 Sprague-Dawley Cd/COPS/SD, rat,

115t HAP/SD, rat, 115t rat, 115t

lesions tail veins lung, 193t Squamous cell carcinoma, 47, 63f, 64f,

65f anterior nasal cavity, 56f lung F344 rat, 129f

hamster, 119f, 132t rat, 124, 125f, 128f

naturally occurring, 126t radiation-induced, 127

syrian hamster, 117 nasal cavity rat, 58f

mucosa,55f rat, 54, 59t

nasoturbinate rat, 57f pulmonary vein C57Bl J mouse,

130f rat lung, 124

mouse hamster man, comparison, 132t

upper respiratory tract syrian hamster, 62

papilloma, 33 tumor, 62, 117

cells, malignant, 64f, 65f, 118f epithelium, dysplastic, 63f metaplasia, 29f, 30f, 34, 36

alveolar, 199f lung hamster, 118f

Stage, predifferentiation, 11 Staphylococci, 221 Strain, A mouse, 148t, 148t

AlBrA mouse, 148t B6C3HFl mouse, 148t BALB/C/C3H/He mouse, 148t

cfC3H/Cb/Se mouse, 148t cfC3H mouse, 148t cfRIII mouse, 148t cNIV mouse, 148t

C3H/Cb/Se mouse, 148t

Subject Index 239

mouse, 148t C57BLl6J mouse, 148t CBA mouse, 148t Cf-l mouse, 148t DBA mouse, 148t OR mouse, 148t NIH white mouse, 148t RIII/Dm/Se mouse, 148t

mouse, 148t Streptobacillus moniliformis, 215, 221 Streptococcus pneumoniae, 215

rat, 201 Structure function lung, 89 Subcutaneous mass, rat, 55f

sarcoma, lung mouse metastasis, 141f

Submaxillary gland virus infection, rat, 84,210

Sulfonic acid, HEPES Hydroxyethyl piperazineethane, 28t

Sulfoxide, DMSO Dimethyl, 28t Swiss, mice, 106t Syncytia, bronchiole multiple

epithelial, 196f Syncytial virus RSV, respiratory, 209 Syrian golden hamster, carcinogens respiratory epithelium, 33

tracheal epithelium, 11 hamsters, explant cultures fetal trachea, 27

hamster, clear cell carcinoma larynx, 75

pleural mesothelioma, 133 radioactive materials, lung tumor,

132t squamous cell carcinoma lung,

117 upper respiratory tra, 62

System, upper respiratory, 1

Tachyzoites, toxoplasma, 227 Tail veins bleeding, 188t, 193t

hair thromboarteritis, 188t vessel, 193t

lung, lesions, 188t Sprague-Dawley rat, lesions,

193t periphlebitis, 188t, 193t phlebitis, 188t thrombophlebitis hair, 193t

Terreus pleura rat, Aspergillus, 225f Thrombi malignant schwan noma, lung

mouse, 146f Thromboarteritis hair particle, lung,

193t lung, 188t, 193t skin particle, lung, 193t tail veins hair, 188t

Thrombophlebitis hair, tail veins, 193t Thrombotic pulmonary artery, 187f Thrombus caudal vein male rat, 190f Tissue rat, normal bronchial lymphoid,

215f Torulopsis glabrata, Candida, 219,

222f

Page 250: Respiratory System

240 Subject Index

Toxicity bleomycin baboon, pulmonary, 165t

dog, pulmonary, 165t hamster, pulmonary, 165t mouse, pulmonary, 165t pheasant, pulmonary, 165t

Toxoplasma gondii, 227, 229 mouse, 201 tachyzoites, 227

Toxoplasmic pneumonia hamster, 228f nude mouse, 228f

Toxoplasmosis lung mouse hamster, 227

transmission, 229 Trachea, 11f

dysplasia, 35f epithelium rat, 89f explant culture fetal, 28

fetal,30f fetal,30f hamster, 22f

papillary mucoepidermoid tumor, 33f

hyperplasia, 35f normal rat, 81f papillary tumor, 37f seromucous glands rat, 91f syrian golden hamsters, explant

cultures fetal, 27 Tracheal epithelial cell Mycoplasma

pulmonis, 82f epithelium, 13f, 15f, 21f, 24f

hamster, 11f syrian golden hamster, 11

explant, fetal, 29f mucosa,81f

Tract rat, murine respiratory myco­plasmosis upper respiratory, 78

sialodacryoadenitis virus infection upper respirat, 84

Transitional cell carcinoma, lung mouse metastasis renal, 145f

Transmission toxoplasmosis, 229 Trimethoxycinnamaldehyde, 59t Trimethylaniline, 101 t Trophozoites, pneumocystis, 222f Trypanosoma cruzi, 227 Tumor emboli pulmonary artery, lung

mouse mammary, 141f epidermoid papillary, 33 lung mouse metastasis ovarian

granulosa cell, 144f RIll mouse metastatic mammary, 140f

mammary,138f mice radioactive materials, lung,

131t

muco-epidermoid papillary, 36f mucoepidermoid papillary, 33

respiratory, 39f olfactory region, 51f polypoid, 33 rat, lung, 131t squamous cell, 62, 117 syrian hamster radioactive materials,

lung,132t trachea hamster, papillary

mucoepidermoid, 33f papillary, 37f

Tumors, differential diagnosis papillary, 39

F344 rats, chemically induced lung, 10tt

lung mouse, metastatic, 138 secondary, 138

mice, frequency pulmonary, 106t pulmonary metastasis induced, 149t

papillary, 34 rats, naturally occurring bronchiolar alveolar, 115t

ultrastructure, papillary, 37 Turbinate, ethmoid, 4f

rat, ventral, 85f Turbinates rat, neoplasms mucosa

ethmoid,47 Tympanic bulla purulent exudate, 80f Type B, lung mouse metastatic

mammary adenocarcinoma, 143f I cell, 14f, 24f, 25f

mucous cells, 22f II adenoma, 103f

cell, 104f lung mouse, 102f, 103f, 104f

alveolar cell rat, 96f cell, 14f, 19f

adenoma, 102 lung mouse, 105f

alveolar, 102 ciliated cell, 16f, 18f epithelial cell, lung mouse, 163f

Ultrastructure, papillary tumors, 37 Untreated mice, pulmonary metas­tasis, 148t

Upper respiratory system, 1 tract rat, murine respiratory

mycoplasmosis, 78 sialodacryoadenitis virus

infect, 84 syrian hamster, squamous cell

carcino,62 Urinary bladder, metastasis lung, 150t

Vascular lesion bleomycin, lung mouse, 161f system, hair fragment emboli

pulmonary, 186 Vein male rat, thrombus caudal, 190f Veins bleeding, tail, 188t, 193t

hair thromboarteritis, tail, 188t vessel, tail, 193t

lung, lesions tail, 188t Sprague-Dawley, rat, lesions tail,

193t periphlebitis, tail, 188t, 193t phlebitis, tail, 188t thrombophlebitis hair, tail, 193t

VEN N-nitrosovinylethylamine, 28t Ventral turbinate rat, 85f Vessel, tail veins hair, 193t Vessels bleomycin, lung mouse

pulmonary blood, 162f Viral pneumonia, sialodacryo­

adenitis, 211 f Virus infection JHV, Japanese

hemagglutinating, 195 lung mouse rat, sendai, 195

sialodacryoadenitis, 210 mouse HVM, hemagglutinating, 195

parainfluenza, 195 rat submaxillary gland, 84, 210 reparative phase, sendai, 197f resolution phase, sendai, 200f sendai, 196f, 197f upper respiratory tract rat,

sialodacryoadeni, 84 Japan HVJ, hemagglutinating, 195 mice infection lung mouse rat,

pneumonia, 206 pneumonia, 209 PVM, pneumonia, 206 rat lung, pneumonia, 207f

mouse K, 201 pneumonia, 201,206

parainfluenza, 202 pneumonia, 206 RSV, respiratory syncytial, 209 SDA V, sialodacryoadenitis, 210 sialodacryoadenitis, 86

Vomeronasal organ, 4f

White mouse strain, NIH, 148t Wistar, rat, 115t, 126t

Xylidine, 46t

Yeasts, fungal, 222f

Zygomycosis, 224